EP2339014B1 - Procédés et compositions comprenant des acides aminés non naturels - Google Patents

Procédés et compositions comprenant des acides aminés non naturels Download PDF

Info

Publication number
EP2339014B1
EP2339014B1 EP10191428.1A EP10191428A EP2339014B1 EP 2339014 B1 EP2339014 B1 EP 2339014B1 EP 10191428 A EP10191428 A EP 10191428A EP 2339014 B1 EP2339014 B1 EP 2339014B1
Authority
EP
European Patent Office
Prior art keywords
amino acid
natural amino
polypeptide
amino acids
polypeptides
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP10191428.1A
Other languages
German (de)
English (en)
Other versions
EP2339014A1 (fr
Inventor
Zhenwei Miao
Feng Tian
Anna-Maria A. Hays Putnam
Ying Buechler
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ambrx Inc
Original Assignee
Ambrx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ambrx Inc filed Critical Ambrx Inc
Publication of EP2339014A1 publication Critical patent/EP2339014A1/fr
Application granted granted Critical
Publication of EP2339014B1 publication Critical patent/EP2339014B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6806Determination of free amino acids
    • G01N33/6812Assays for specific amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/24Extraction; Separation; Purification by electrochemical means
    • C07K1/26Electrophoresis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/30Extraction; Separation; Purification by precipitation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/34Extraction; Separation; Purification by filtration, ultrafiltration or reverse osmosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/06Preparation of peptides or proteins produced by the hydrolysis of a peptide bond, e.g. hydrolysate products
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B40/00Libraries per se, e.g. arrays, mixtures
    • C40B40/04Libraries containing only organic compounds
    • C40B40/10Libraries containing peptides or polypeptides, or derivatives thereof
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor

Definitions

  • non-genetically encoded amino acids i.e., "non-natural amino acids”
  • chemical functional groups that could provide valuable alternatives to the naturally-occurring functional groups, such as the epsilon-NH2 of lysine, the sulfhydryl -SH of cysteine, the imino group of histidine, etc.
  • Certain chemical functional groups are known to be inert to the functional groups found in the 20 common, genetically-encoded amino acids but react cleanly and efficiently to form stable linkages with functional groups that can be incorporated onto non-natural amino acids.
  • Described herein are methods, compositions, techniques and strategies for making, purifying, detecting, characterizing, and using non-natural amino acids, non-natural amino acid polypeptides and modified non-natural amino acid polypeptides.
  • composition comprising a polypeptide covalently linked to a nucleic acid molecule at one or more specific amino acid positions of said polypeptide, wherein said nucleic acid molecule is covalently linked to an amino acid side chain of a non-naturally encoded amino acid of said polypeptide.
  • a method of detecting a polypeptide that comprises detecting a non-naturally encoded amino acid side chain in the polypeptide.
  • the polypeptide is ribosomally synthesized.
  • methods of detecting a polypeptide that comprise detecting a non-naturally encoded amino acid side chain in the polypeptide that has been post-translationally modified are also disclosed.
  • the method comprises contacting the polypeptide with a substance that interacts with the non-naturally encoded amino acid side chain in the polypeptide.
  • the method of purifying a polypeptide having a non-naturally encoded amino acid in the polypeptide chain comprises precipitation of the polypeptide, wherein the non-naturally encoded amino acid alters the solubility of the polypeptide when compared to the solubility of the polypeptide without a non-naturally encoded amino acid in the polypeptide chain.
  • Methods of purifying a ribosomally made polypeptide having a non-naturally encoded amino acid in the polypeptide side chain comprises electrophoresis of the polypeptide, wherein the non-naturally encoded amino acid alters the electrophoretic mobility of the polypeptide when compared to the electrophoretic mobility of the polypeptide without a non-naturally encoded amino acid in the polypeptide chain are also disclosed.
  • the method of purifying a ribosomally made polypeptide having a non-naturally encoded amino acid in the polypeptide side chain comprises dialysis of the polypeptide, wherein the non-naturally encoded amino acid alters the diffusion rate of the polypeptide when compared to the diffusion rate of the polypeptide without a non-naturally encoded amino acid in the polypeptide chain.
  • Also disclosed herein is a method for screening a library of molecules, comprising: a) combining a polypeptide comprising a non-naturally encoded amino acid with the library molecules under conditions to allow interaction of the library molecules with the polypeptide comprising a non-naturally encoded amino acid, and b) identifying the library molecules which interact with the polypeptide comprising a non-naturally encoded amino acid.
  • a library of ribosomally made polypeptides comprising a plurality of polypeptides having different amino acid sequences, wherein each polypeptide comprises a non-natural amino acid is screened.
  • Also disclosed herein is a method, comprising: a) substituting a non-naturally encoded amino acid for a naturally encoded amino acid at a single pre-selected site in a pre-selected polypeptide having at least one known biological activity; and b) measuring a biological activity of the pre-selected polypeptide comprising the non-naturally encoded amino acid; and c) comparing the biological activity of the pre-selected polypeptide of step b) with the pre-selected polypeptide having a non-naturally encoded amino acid substituted for a naturally encoded amino acid at a different position in the pre-selected polypeptide chain or with the pre-selected polypeptide without a substituted non-naturally encoded amino acid in the polypeptide chain.
  • a method for selecting a position for post-translational modification of a pre-selected polypeptide comprises a) substituting a non-naturally encoded amino acid for a naturally encoded amino acid at a single pre-selected site in a pre-selected polypeptide having at least one known biological activity; and b) measuring a biological activity of the pre-selected polypeptide comprising the non-naturally encoded amino acid; and c) comparing the biological activity of the pre-selected polypeptide of step b) with the pre-selected polypeptide having a non-naturally encoded amino acid substituted for a naturally encoded amino acid at a different position in the pre-selected polypeptide chain or with the pre-selected polypeptide without a substituted non-naturally encoded amino acid in the polypeptide chain.
  • alkoxy alkylamino and “alkylthio” (or thioalkoxy) are used in their conventional sense, and refer to those alkyl groups attached to the remainder of the molecule via an oxygen atom, an amino group, or a sulfur atom, respectively.
  • alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain, or cyclic hydrocarbon radical, or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di- and multivalent radicals, having the number of carbon atoms designated ( i.e. C 1 -C 10 means one to ten carbons).
  • saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • An unsaturated alkyl group is one having one or more double bonds or triple bonds.
  • alkyl groups examples include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
  • alkyl unless otherwise noted, is also meant to include those derivatives of alkyl defined in more detail below, such as “heteroalkyl.”
  • Alkyl groups which are limited to hydrocarbon groups are termed "homoalkyl".
  • alkylene by itself or as part of another substituent means a divalent radical derived from an alkane, as exemplified, but not limited, by the structures -CH 2 CH 2 - and -CH 2 CH 2 CH 2 CH 2 -, and further includes those groups described below as “heteroalkylene.”
  • an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being a particular embodiment of the methods and compositions described herein.
  • a “lower alkyl,” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
  • amino acid refers to naturally occurring and non-natural amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally encoded amino acids are the 20 common amino acids (alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine) and pyrrolysine and selenocysteine.
  • Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an ⁇ carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, such as, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium.
  • Such analogs have modified R groups (such as, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
  • amino terminus modification group refers to any molecule that can be attached to the amino terminus of a polypeptide.
  • a “carboxy terminus modification group” refers to any molecule that can be attached to the carboxy terminus of a polypeptide.
  • Terminus modification groups include but are not limited to various water soluble polymers, peptides or proteins such as serum albumin, or other moieties that increase serum half-life of peptides.
  • aryl means, unless otherwise stated, a polyunsaturated, aromatic, hydrocarbon substituent which can be a single ring or multiple rings (including but not limited to, from 1 to 3 rings) which are fused together or linked covalently.
  • heteroaryl refers to aryl groups (or rings) that contain from one to four heteroatoms selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • Non-limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinoly
  • aryl when used in combination with other terms (including but not limited to, aryloxy, arylthioxy, aralkyl) includes both aryl and heteroaryl rings as defined above.
  • aralkyl or “alkaryl” is meant to include those radicals in which an aryl group is attached to an alkyl group (including but not limited to, benzyl, phenethyl, pyridylmethyl and the like) including those alkyl groups in which a carbon atom (including but not limited to, a methylene group) has been replaced by, for example, an oxygen atom (including but not limited to, phenoxymethyl, 2-pyridyloxymethyl, 3-(1-naphthyloxy)propyl, and the like).
  • a “bifunctional polymer” refers to a polymer comprising two discrete functional groups that are capable of reacting specifically with other moieties (including but not limited to, amino acid side groups) to form covalent or non-covalent linkages.
  • a bifunctional linker having one functional group reactive with a group on a particular biologically active component, and another group reactive with a group on a second biological component may be used to form a conjugate that includes the first biologically active component, the bifunctional linker and the second biologically active component.
  • Many procedures and linker molecules for attachment of various compounds to peptides are known. See, e.g., European Patent Application No. 188,256 ; U.S. Patent Nos.
  • a "multi-functional polymer” refers to a polymer comprising two or more discrete functional groups that are capable of reacting specifically with other moieties (including but not limited to, amino acid side groups) to form covalent or non-covalent linkages.
  • a bi-functional polymer or multi-functional polymer may be any desired length or molecular weight, and may be selected to provide a particular desired spacing or conformation between one or more molecules linked to the polypeptide and its binding partner or the polypeptide.
  • biologically active molecule means any substance which can affect any physical or biochemical properties of a biological system, pathway, molecule, or interaction relating to an organism, including but not limited to viruses, bacteria, bacteriophage, transposon, prion, insects, fungi, plants, animals, and humans.
  • biologically active molecules include but are not limited to any substance intended for diagnosis, cure, mitigation, treatment, or prevention of disease in humans or other animals, or to otherwise enhance physical or mental well-being of humans or animals.
  • biologically active molecules include, but are not limited to, peptides, proteins, enzymes, small molecule drugs, hard drugs, soft drugs, carbohydrates, inorganic atoms or molecules, dyes, lipids, nucleosides, radionuclides, oligonucleotides, toxins, cells, viruses, liposomes, microparticles and micelles.
  • Classes of biologically active agents that are suitable for use with the methods and compositions described herein include, but are not limited to, drugs, prodrugs, radionuclides, imaging agents, polymers, antibiotics, fungicides, anti-viral agents, anti-inflammatory agents, anti-tumor agents, cardiovascular agents, anti-anxiety agents, hormones, growth factors, steroidal agents, microbially derived toxins, and the like.
  • Cofolding refers specifically to refolding processes, reactions, or methods which employ at least two polypeptides which interact with each other and result in the transformation of unfolded or improperly folded polypeptides to native, properly folded polypeptides.
  • a “comparison window,” as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Methods of alignment of sequences for comparison are well-known in the art.
  • Optimal alignment of sequences for comparison can be conducted, including but not limited to, by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math. 2:482c , by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol.
  • BLAST and BLAST 2.0 algorithms are described in Altschul et al. (1997) Nuc. Acids Res. 25:3389-3402 , and Altschul et al. (1990) J. Mol. Biol. 215:403-410 , respectively.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • the BLAST algorithm is typically performed with the "low complexity" filter turned off.
  • the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5787 ).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, less than about 0.01, or less than about 0.001.
  • “conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, “conservatively modified variants” refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide.
  • nucleic acid variations are "silent variations," which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
  • each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan
  • TGG which is ordinarily the only codon for tryptophan
  • amino acid sequences one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are known to those of ordinary skill in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the methods and compositions described herein.
  • cycloalkyl and heterocycloalkyl represent, unless otherwise stated, cyclic versions of “alkyl” and “heteroalkyl”, respectively.
  • a cycloalkyl or heterocycloalkyl include saturated, partially unsaturated and fully unsaturated ring linkages.
  • a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule.
  • Examples of cycloalkyl include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
  • heterocycloalkyl examples include, but are not limited to, 1-(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, and the like. Additionally, the term encompasses bicyclic and tricyclic ring structures.
  • heterocycloalkylene by itself or as part of another substituent means a divalent radical derived from heterocycloalkyl
  • cycloalkylene by itself or as part of another substituent means a divalent radical derived from cycloalkyl
  • Denaturing agent or “denaturant,” as used herein, is defined as any compound or material which will cause a reversible unfolding of a protein.
  • the strength of a denaturing agent or denaturant will be determined both by the properties and the concentration of the particular denaturing agent or denaturant.
  • Suitable denaturing agents or denaturants may be chaotropes, detergents, organic, water miscible solvents, phospholipids, or a combination of two or more such agents. Suitable chaotropes include, but are not limited to, urea, guanidine, and sodium thiocyanate.
  • Useful detergents may include, but are not limited to, strong detergents such as sodium dodecyl sulfate, or polyoxyethylene ethers (e.g. Tween or Triton detergents), Sarkosyl, mild non-ionic detergents (e.g. , digitonin), mild cationic detergents such as N->2,3-(Dioleyoxy)-propyl-N,N,N-trimethylammonium, mild ionic detergents (e.g.
  • sodium cholate or sodium deoxycholate or zwitterionic detergents including, but not limited to, sulfobetaines (Zwittergent), 3-(3-chlolamidopropyl)dimethylammonio-1-propane sulfate (CHAPS), and 3-(3-chlolamidopropyl)dimethylammonio-2-hydroxy-1-propane sulfonate (CHAPSO).
  • Zwittergent 3-(3-chlolamidopropyl)dimethylammonio-1-propane sulfate
  • CHAPSO 3-(3-chlolamidopropyl)dimethylammonio-2-hydroxy-1-propane sulfonate
  • Organic, water miscible solvents such as acetonitrile, lower alkanols (especially C 2 - C 4 alkanols such as ethanol or isopropanol), or lower alkandiols (especially C 2 - C 4 alkandiols such as ethyleneglycol) may be used as denaturants.
  • lower alkanols especially C 2 - C 4 alkanols such as ethanol or isopropanol
  • lower alkandiols especially C 2 - C 4 alkandiols such as ethyleneglycol
  • Phospholipids useful in the methods and compositions described herein may be naturally occurring phospholipids such as phosphatidylethanolamine, phosphatidylcholine, phosphatidylserine, and phosphatidylinositol or synthetic phospholipid derivatives or variants such as dihexanoylphosphatidylcholine or diheptanoylphosphatidylcholine.
  • naturally occurring phospholipids such as phosphatidylethanolamine, phosphatidylcholine, phosphatidylserine, and phosphatidylinositol or synthetic phospholipid derivatives or variants such as dihexanoylphosphatidylcholine or diheptanoylphosphatidylcholine.
  • compositions containing the (modified) non-natural amino acid polypeptide described herein can be administered for prophylactic, enhancing, and/or therapeutic treatments.
  • an “enhance” or “enhancing” means to increase or prolong either in potency or duration a desired effect.
  • the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system.
  • An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system. When used in a patient, amounts effective for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician.
  • the term "eukaryote” refers to organisms belonging to the phylogenetic domain Eucarya such as animals (including but not limited to, mammals, insects, reptiles, birds, etc. ), ciliates, plants (including but not limited to, monocots, dicots, algae, etc. ), fungi, yeasts, flagellates, microsporidia, protists, etc.
  • halogen includes fluorine, chlorine, iodine, and bromine.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and at least one heteroatom selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule.
  • heteroalkylene by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified, but not limited by, -CH 2 -CH 2 -S-CH 2 -CH 2 - and -CH 2 -S-CH 2 -CH 2 -NH-CH 2 -.
  • heteroalkylene groups the same or different heteroatoms can also occupy either or both of the chain termini (including but not limited to, alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, aminooxyalkylene, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied by the direction in which the formula of the linking group is written. For example, the formula-C(O) 2 R'- represents both -C(O) 2 R'- and -R'C(O) 2 -.
  • nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same. Sequences are “substantially identical” if they have a percentage of amino acid residues or nucleotides that are the same (i.e., about 60% identity, optionally about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95% identity over a specified region), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. This definition also refers to the complement of a test sequence.
  • the identity can exist over a region that is at least about 50 amino acids or nucleotides in length, or over a region that is 75-100 amino acids or nucleotides in length, or, where not specified, across the entire sequence of a polynucleotide or polypeptide.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • isolated when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is free of at least some of the cellular components with which it is associated in the natural state, or that the nucleic acid or protein has been concentrated to a level greater than the concentration of its in vivo or in vitro production. It can be in a homogeneous state. Isolated substances can be in either a dry or semi-dry state, or in solution, including but not limited to an aqueous solution. It can be a component of a pharmaceutical composition that comprises additional pharmaceutically acceptable carriers and/or excipients. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography.
  • a protein which is the predominant species present in a preparation is substantially purified.
  • an isolated gene is separated from open reading frames which flank the gene and encode a protein other than the gene of interest.
  • the term "purified” denotes that a nucleic acid or protein gives rise to substantially one band in an electrophoretic gel. Particularly, it may mean that the nucleic acid or protein is at least 85% pure, at least 90% pure, at least 95% pure, at least 99% or greater pure.
  • linkage or “linker” is used herein to refer to groups or bonds that normally are formed as the result of a chemical reaction and typically are covalent linkages.
  • Hydrolytically stable linkages means that the linkages are substantially stable in water and do not react with water at useful pH values, including but not limited to, under physiological conditions for an extended period of time, perhaps even indefinitely.
  • Hydrolytically unstable or degradable linkages mean that the linkages are degradable in water or in aqueous solutions, including for example, blood.
  • Enzymatically unstable or degradable linkages mean that the linkage can be degraded by one or more enzymes.
  • PEG and related polymers may include degradable linkages in the polymer backbone or in the linker group between the polymer backbone and one or more of the terminal functional groups of the polymer molecule.
  • ester linkages formed by the reaction of PEG carboxylic acids or activated PEG carboxylic acids with alcohol groups on a biologically active agent generally hydrolyze under physiological conditions to release the agent.
  • hydrolytically degradable linkages include but are not limited to carbonate linkages; imine linkages resulted from reaction of an amine and an aldehyde; phosphate ester linkages formed by reacting an alcohol with a phosphate group; hydrazone linkages which are reaction product of a hydrazide and an aldehyde; acetal linkages that are the reaction product of an aldehyde and an alcohol; orthoester linkages that are the reaction product of a formate and an alcohol; peptide linkages formed by an amine group, including but not limited to, at an end of a polymer such as PEG, and a carboxyl group of a peptide; and oligonucleotide linkages formed by a phosphoramidite group, including but not limited to, at the end of a polymer, and a 5' hydroxyl group of an oligonucleotide.
  • the term “medium” or “media” includes any culture medium, solution, solid, semi-solid, or rigid support that may support or contain any host cell, including bacterial host cells, yeast host cells, insect host cells, plant host cells, eukaryotic host cells, mammalian host cells, CHO cells, prokaryotic host cells, E. coli, or Pseudomonas host cells, and cell contents.
  • the term may encompass medium in which the host cell has been grown, e.g. , medium into which the polypeptide has been secreted, including medium either before or after a proliferation step.
  • the term also may encompass buffers or reagents that contain host cell lysates, such as in the case where the polypeptide is produced intracellularly and the host cells are lysed or disrupted to release the polypeptide.
  • a “metabolite” of a (modified) non-natural amino acid polypeptide disclosed herein is a derivative of that (modified) non-natural amino acid polypeptide that is formed when the (modified) non-natural amino acid polypeptide is metabolized.
  • active metabolite refers to a biologically active derivative of a (modified) non-natural amino acid polypeptide that is formed when the (modified) non-natural amino acid polypeptide is metabolized.
  • the term “metabolized” refers to the sum of the processes (including, but not limited to, hydrolysis reactions and reactions catalyzed by enzymes) by which a particular substance is changed by an organism.
  • Metabolites of the (modified) non-natural amino acid polypeptide disclosed herein can be identified either by administration of (modified) non-natural amino acid polypeptide to a host and analysis of tissue samples from the host, or by incubation of (modified) non-natural amino acid polypeptide with hepatic cells in vitro and analysis of the resulting compounds.
  • modified refers to the presence of a post-translational modification on a polypeptide.
  • the form “(modified)” term means that the polypeptides being discussed are optionally modified, that is, the polypeptides under discussion can be modified or unmodified.
  • modulated serum half-life means the positive or negative change in circulating half-life of a (modified) polypeptide relative to its non-modified form. Serum half-life is measured by taking blood samples at various time points after administration of the polypeptide, and determining the concentration of that molecule in each sample. Correlation of the serum concentration with time allows calculation of the serum half-life. Increased serum half-life desirably has at least about two-fold, but a smaller increase may be useful, for example where it enables a satisfactory dosing regimen or avoids a toxic effect. In some embodiments, the increase is at least about three-fold, at least about five-fold, or at least about ten-fold.
  • modulated therapeutic half-life means the positive or negative change in the half-life of the therapeutically effective amount of a (modified) polypeptide, relative to its non-modified form.
  • Therapeutic half-life is measured by measuring pharmacokinetic and/or pharmacodynamic properties of the molecule at various time points after administration. Increased therapeutic half-life desirably enables a particular beneficial dosing regimen, a particular beneficial total dose, or avoids an undesired effect. In some embodiments, the increased therapeutic half-life results from increased potency, increased or decreased binding of the modified molecule to its target, incrased or decreased breakdown of the molecule by enzymes such as proteases, or an increase or decrease in another parameter or mechanism of action of the non-modified molecule.
  • non-eukaryote refers to non-eukaryotic organisms.
  • a non-eukaryotic organism can belong to the Eubacteria (including but not limited to, Escherichia coli, Thermus thermophilus, Bacillus stearothermophilus, Pseudomonas fluorescens, Pseudomonas aeruginosa, Pseudomonas putida, etc.
  • phylogenetic domain or the Archaea (including but not limited to, Methanococcus jannaschii, Methanobacterium thermoautotrophicum, Halobacterium such as Haloferax volcanii and Halobacterium species NRC-1, Archaeoglobus fulgidus, Pyrococcus furiosus, Pyrococcus horikoshii, Aeuropyrum pernix, etc .) phylogenetic domain.
  • Archaea including but not limited to, Methanococcus jannaschii, Methanobacterium thermoautotrophicum, Halobacterium such as Haloferax volcanii and Halobacterium species NRC-1, Archaeoglobus fulgidus, Pyrococcus furiosus, Pyrococcus horikoshii, Aeuropyrum pernix, etc .
  • non-natural amino acid refers to an amino acid that is not one of the 20 common amino acids or pyrrolysine or selenocysteine; other terms that may be used synonymously with the term “non-natural amino acid” is “non-naturally encoded amino acid,” “unnatural amino acid,” “non-naturally-occurring amino acid,” and variously hyphenated and non-hyphenated versions thereof.
  • non-natural amino acid includes, but is not limited to, amino acids that occur naturally by modification of a naturally encoded amino acid (including but not limited to, the 20 common amino acids or pyrrolysine and selenocysteine) but are not themselves incorporated into a growing polypeptide chain by the translation complex.
  • Naturally-occurring amino acids that are not naturally-encoded include, but are not limited to, N -acetylglucosaminyl-L-serine, N -acetylglucosaminyl-L-threonine, and O-phosphotyrosine.
  • nucleic acid refers to deoxyribonucleotides, deoxyribonucleosides, ribonucleosides or ribonucleotides and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless specifically limited otherwise, the term also refers oligonucleotide analogs including PNA (peptidonucleic acid), analogs of DNA used in antisense technology (phosphorothioates, phosphoroamidates, and the like).
  • PNA peptidonucleic acid
  • analogs of DNA used in antisense technology phosphorothioates, phosphoroamidates, and the like.
  • nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (including but not limited to, degenerate codon substitutions) and complementary sequences as well as the sequence explicitly indicated.
  • degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues ( Batzer et al., Nucleic Acid Res. 19:5081 (1991 ); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985 ); and Rossolini et al., Mol. Cell. Probes 8:91-98 (1994 )).
  • Oxidizing agent as used herein with respect to protein refolding, is defined as any compound or material which is capable of removing an electron from a compound being oxidized. Suitable oxidizing agents include, but are not limited to, oxidized glutathione, cystine, cystamine, oxidized dithiothreitol, oxidized erythreitol, and oxygen. A wide variety of oxidizing agents are suitable for use in the methods and compositions described herein.
  • polyalkylene glycol refers to polyethylene glycol, polypropylene glycol, polybutylene glycol, and derivatives thereof.
  • polyalkylene glycol encompasses both linear and branched polymers and average molecular weights of between 1 kDa and 100 kDa.
  • Other exemplary embodiments are listed, for example, in commercial supplier catalogs, such as Shearwater Corporation's catalog “Polyethylene Glycol and Derivatives for Biomedical Applications” (2001 ).
  • polypeptide peptide
  • protein protein
  • polypeptide peptide
  • peptide protein
  • the terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-natural amino acid.
  • the terms encompass amino acid chains of any length, including full length proteins, wherein the amino acid residues are linked by covalent peptide bonds.
  • post-translationally modified refers to any modification of a natural or non-natural amino acid that occurs to such an amino acid after it has been incorporated into a polypeptide chain.
  • the term encompasses, by way of example only, co-translational in vivo modifications, co-translational in vitro modifications (such as in a cell-free translation system), post-translational in vivo modifications, and post-translational in vitro modifications.
  • prodrug refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • compositions containing the (modified) non-natural amino acid polypeptide are administered to a patient susceptible to or otherwise at risk of a particular disease, disorder or condition.
  • a patient susceptible to or otherwise at risk of a particular disease, disorder or condition is defined to be a "prophylactically effective amount.”
  • prophylactically effective amounts In this use, the precise amounts also depend on the patient's state of health, weight, and the like. It is considered well within the skill of the art for one to determine such prophylactically effective amounts by routine experimentation ( e.g. , a dose escalation clinical trial).
  • the term "protected” refers to the presence of a “protecting group” or moiety that prevents reaction of the chemically reactive functional group under certain reaction conditions.
  • the protecting group will vary depending on the type of chemically reactive group being protected. For example, if the chemically reactive group is an amine or a hydrazide, the protecting group can be selected from the group of tert-butyloxycarbonyl (t-Boc) and 9-fluorenylmethoxycarbonyl (Fmoc). If the chemically reactive group is a thiol, the protecting group can be orthopyridyldisulfide.
  • the chemically reactive group is a carboxylic acid, such as butanoic or propionic acid, or a hydroxyl group
  • the protecting group can be benzyl or an alkyl group such as methyl, ethyl, or tert-butyl.
  • Other protecting groups known in the art may also be used in or with the methods and compositions described herein, including photolabile groups such as Nvoc and MeNvoc.
  • blocking/protecting groups may be selected from:
  • a “recombinant host cell” or “host cell” refers to a cell that includes an exogenous polynucleotide, regardless of the method used for insertion, for example, direct uptake, transduction, f-mating, or other methods known in the art to create recombinant host cells.
  • the exogenous polynucleotide may be maintained as a nonintegrated vector, for example, a plasmid, or alternatively, may be integrated into the host genome.
  • Reducing agent as used herein with respect to protein refolding, is defined as any compound or material which maintains sulfhydryl groups in the reduced state and reduces intra- or intermolecular disulfide bonds.
  • Suitable reducing agents include, but are not limited to, dithiothreitol (DTT), 2-mercaptoethanol, dithioerythritol, cysteine, cysteamine (2-aminoethanethiol), and reduced glutathione.
  • DTT dithiothreitol
  • 2-mercaptoethanol dithioerythritol
  • cysteine cysteine
  • cysteamine (2-aminoethanethiol) 2-aminoethanethiol
  • reduced glutathione A wide variety of reducing agents are suitable for use in the methods and compositions described herein.
  • Refolding describes any process, reaction or method which transforms disulfide bond containing polypeptides from an improperly folded or unfolded state to a native or properly folded conformation with respect to disulfide bonds.
  • the phrase "selectively (or specifically) hybridizes to” refers to the binding, duplexing, or hybridizing of a molecule only to a particular nucleotide sequence under stringent hybridization conditions when that sequence is present in a complex mixture (including but not limited to, total cellular or library DNA or RNA).
  • stringent hybridization conditions refers to conditions of low ionic strength and high temperature as is known in the art. Typically, under stringent conditions a probe will hybridize to its target subsequence in a complex mixture of nucleic acid (including but not limited to, total cellular or library DNA or RNA) but does not hybridize to other sequences in the complex mixture. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Laboratory Techniques in Biochemistry and Molecular Biology--Hybridization with Nucleic Probes, "Overview of principles of hybridization and the strategy of nucleic acid assays" (1993 ).
  • stringent conditions are selected to be about 5-10° C lower than the thermal melting point (T m ) for the specific sequence at a defined ionic strength pH.
  • T m is the temperature (under defined ionic strength, pH, and nucleic concentration) at which 50% of the probes complementary to the target hybridize to the target sequence at equilibrium (as the target sequences are present in excess, at T m , 50% of the probes are occupied at equilibrium).
  • Stringent conditions may be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30°C for short probes (including but not limited to, 10 to 50 nucleotides) and at least about 60° C for long probes (including but not limited to, greater than 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide. For selective or specific hybridization, a positive signal may be at least two times background, optionally 10 times background hybridization.
  • Exemplary stringent hybridization conditions can be as following: 50% formamide, 5X SSC, and 1% SDS, incubating at 42°C, or 5X SSC, 1% SDS, incubating at 65°C, with wash in 0.2X SSC, and 0.1% SDS at 65°C. Such washes can be performed for 5, 15, 30, 60, 120, or more minutes.
  • subject refers to an animal, in some embodiments a mammal, and in other embodiments a human, who is the object of treatment, observation or experiment.
  • substantially purified refers to a polypeptide that may be substantially or essentially free of components that normally accompany or interact with the protein as found in its naturally occurring environment, i.e. a native cell, or host cell in the case of recombinantly produced polypeptide.
  • a polypeptide that may be substantially free of cellular material includes preparations of protein having less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% (by dry weight) of contaminating protein.
  • the protein may be present at about 30%, about 25%, about 20%, about 15%, about 10%, about 5%, about 4%, about 3%, about 2%, or about 1% or less of the dry weight of the cells.
  • the protein may be present in the culture medium at about 5g/L, about 4g/L, about 3g/L, about 2g/L, about 1g/L, about 750mg/L, about 500mg/L, about 250mg/L, about 100mg/L, about 50mg/L, about 10mg/L, or about 1mg/L or less of the dry weight of the cells.
  • substantially purified polypeptide as produced by the methods described herein may have a purity level of at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, specifically, a purity level of at least about 75%, 80%, 85%, and more specifically, a purity level of at least about 90%, a purity level of at least about 95%, a purity level of at least about 99% or greater as determined by appropriate methods such as SDS/PAGE analysis, RP-HPLC, SEC, and capillary electrophoresis.
  • non-interfering substituents include but is not limited to "non-interfering substituents.”
  • “Non-interfering substituents” are those groups that yield stable compounds. Suitable non-interfering substituents or radicals include, but are not limited to, halo, C 1 -C 10 alkyl, C 2 -C 10 alkenyl, C 2 -C 10 alkynyl, C 1 -C 10 alkoxy, C 5 -C 12 aralkyl, C 3 -C 12 cycloalkyl, C 4 -C 12 cycloalkenyl, phenyl, substituted phenyl, toluoyl, xylenyl, biphenyl, C 2 -C 12 alkoxyalkyl, C 5 -C 12 alkoxyaryl, C 5 -C 12 aryloxyalkyl, C 7 -C 12 oxyaryl, C 1 -C 6 alkylsulfinyl, C 1
  • R group in the preceding list is independently selected from the group consisting of H, alkyl or substituted alkyl, aryl or substituted aryl, or alkaryl.
  • substituent groups are specified by their conventional chemical formulas, written from left to right, they equally encompass the chemically identical substituents that would result from writing the structure from right to left, for example, -CH 2 O- is equivalent to -OCH 2 -.
  • Each R group in the preceding list is independently selected from the group consisting of hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, including but not limited to, aryl substituted with 1-3 halogens, substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or aralkyl groups.
  • aryl substituted with 1-3 halogens substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or aralkyl groups.
  • -NR 2 is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl.
  • alkyl is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (including but not limited to, -CF 3 and -CH 2 CF 3 ) and acyl (including but not limited to, -C(O)CH 3 , -C(O)CF 3 , -C(O)CH 2 OCH 3 , and the like).
  • compositions containing the (modified) non-natural amino acid polypeptide are administered to a patient already suffering from a disease, condition or disorder, in an amount sufficient to cure or at least partially arrest the symptoms of the disease, disorder or condition.
  • an amount is defined to be a "therapeutically effective amount,” and will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician. It is considered well within the skill of the art for one to determine such therapeutically effective amounts by routine experimentation ( e.g. , a dose escalation clinical trial).
  • test ligand refers to an agent, which can be a compound, molecule or complex, which is being tested for its ability to bind to a non-natural amino acid polypeptide, such as a protein or-protein complex in its native form is known to be associated with or causative of a disease or condition in a living organism, such as a vertebrate, particularly a mammal and even more particularly a human. Since binding of a ligand to its non-natural amino acid polypeptide must occur for the ligand to have a direct effect on the non-natural amino acid polypeptide, binding as indicated by the present assay method is a strong indication of the therapeutic potential of a ligand identified as described herein.
  • a test ligand which can be assessed by the present method can be virtually any agent, including, but not limited to, metals, polypeptides, proteins, lipids, polysaccharides, polynucleotides and small organic molecules.
  • a test ligand which is shown to bind a non-natural amino acid polypeptide is referred to as a ligand.
  • Complex mixtures of substances, including but not limited to, natural product extracts, which include more than one test ligand can be tested and if there is a positive response ( i.e., if binding to the non-natural amino acid polypeptide occurs), the ligand which bound the non-natural amino acid polypeptide can be purified from the mixture prior to further assessment of its therapeutic potential.
  • treating is used to refer to either prophylactic and/or therapeutic treatments.
  • water soluble polymer refers to any polymer that is soluble in aqueous solvents. Linkage of water soluble polymers to a polypeptide can result in changes including, but not limited to, increased or modulated serum half-life, or increased or modulated therapeutic half-life relative to the unmodified form, modulated immunogenicity, modulated physical association characteristics such as aggregation and multimer formation, altered receptor binding, altered binding to one or more binding partners, and altered receptor dimerization or multimerization.
  • the water soluble polymer may or may not have its own biological activity and may be utilized as a linker for attaching the polypeptide to other substances, including but not limited to one or more polypeptides, or one or more biologically active molecules.
  • Suitable polymers include, but are not limited to, polyethylene glycol, polyethylene glycol propionaldehyde, mono C1-C10 alkoxy or aryloxy derivatives thereof (described in U.S. Patent No. 5,252,714 ), monomethoxy-polyethylene glycol, polyvinyl pyrrolidone, polyvinyl alcohol, polyamino acids, divinylether maleic anhydride, N -(2-Hydroxypropyl)-methacrylamide, dextran, dextran derivatives including dextran sulfate, polypropylene glycol, polypropylene oxide/ethylene oxide copolymer, polyoxyethylated polyol, heparin, heparin fragments, polysaccharides, oligosaccharides, glycans, cellulose and cellulose derivatives, including but not limited to methylcellulose and carboxymethyl cellulose, starch and starch derivatives, polypeptides, polyalkylene glycol and derivatives
  • Compounds (including, but not limited to non-natural amino acids, (modified) non-natural amino acid polypeptides and reagents for producing either of the aforementioned compounds) presented herein include isotopically-labelled compounds, which are identical to those recited in the various formulas and structures presented herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into the present compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, such as 2 H, 3 H. 13 C, 14 C, 15 N, 18 O, 17 O, 35 S, 18 F , 36 Cl, respectively.
  • isotopically-labelled compounds described herein for example those into which radioactive isotopes such as 3 H and 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. Further, substitution with isotopes such as deuterium, i.e., 2 H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements.
  • Some of the compounds herein have asymmetric carbon atoms and can therefore exist as enantiomers or diastereomers.
  • Diasteromeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known, for example, by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g. , alcohol), separating the diastereomers and converting ( e.g. , hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. All such isomers, including diastereomers, enantiomers, and mixtures thereof are considered as part of the compositions described herein.
  • the compounds described herein are used in the form of prodrugs.
  • the compounds described herein including, but not limited to non-natural amino acids, (modified) non-natural amino acid polypeptides and reagents for producing either of the aforementioned compounds
  • non-natural amino acids and (modified) non-natural amino acid polypeptides may exist as tautomers. All tautomers are included within the scope of the non-natural amino acids and (modified) non-natural amino acid polypeptides presented herein.
  • the non-natural amino acids and (modified) non-natural amino acid polypeptides described herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms of the non-natural amino acids and (modified) non-natural amino acid polypeptides presented herein are also considered to be disclosed herein.
  • Some of the compounds herein are acidic and may form a salt with a pharmaceutically acceptable cation. Some of the compounds herein (including, but not limited to non-natural amino acids, (modified) non-natural amino acid polypeptides and reagents for producing either of the aforementioned compounds) can be basic and accordingly, may form a salt with a pharmaceutically acceptable anion. All such salts, including di-salts are within the scope of the compositions described herein and they can be prepared by conventional methods.
  • salts can be prepared by contacting the acidic and basic entities, in either an aqueous, nonaqueous or partially aqueous medium.
  • the salts are recovered by using at least one of the following techniques: filtration, precipitation with a non-solvent followed by filtration, evaporation of the solvent, or, in the case of aqueous solutions, lyophilization.
  • Salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene-1-carboxylic acid,
  • Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
  • Acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.
  • a reference to a salt includes the solvent addition forms or crystal forms thereof, particularly solvates or polymorphs.
  • Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are often formed during the process of crystallization. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol.
  • Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and solubility. Various factors such as the recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate.
  • Figure 1 is an overview of the compositions, methods and techniques that are described herein.
  • 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 are the tools (methods, compositions, techniques) for creating and using a polypeptide comprising at least one non-natural amino acid or modified non-natural amino acid.
  • Such non-natural amino acid polypeptides may contain further functionality, including but not limited to, a label; a dye; a polymer; a water-soluble polymer; a derivative of polyethylene glycol; a photocrosslinker; a cytotoxic compound; a drug; an affinity label; a photoaffinity label; a reactive compound; a resin; a second protein or polypeptide or polypeptide analog; an antibody or antibody fragment; a metal chelator; a cofactor; a fatty acid; a carbohydrate; a polynucleotide; a DNA; a RNA; an antisense polynucleotide; a saccharide, a water-soluble dendrimer, a cyclodextrin, an inhibitory ribonucleic acid; a biomaterial; a nanoparticle; a spin label; a fluorophore, a metal-containing moiety; a radioactive moiety; a novel functional group; a group that covalently or
  • the new polypeptide may be designed de novo, including by way of example only, as part of high-throughput screening process (in which case numerous polypeptides may be designed, synthesized, characterized and/or tested) or based on the interests of the researcher.
  • the new polypeptide may also be designed based on the structure of a known or partially characterized polypeptide.
  • the Growth Hormone Gene Superfamily has been the subject of intense study by the scientific community; a new polypeptide may be designed based on the structure of a member or members of this gene superfamily.
  • the principles for selecting which amino acid(s) to substitute and/or modify are described separately herein.
  • the choice of which modification to employ is also described herein, and can be used to meet the need of the experimenter or end user.
  • Modifications include, by way of example only, manipulating the therapeutic effectiveness of the polypeptide, improving the safety profile of the polypeptide, adjusting the pharmacokinetics of the polypeptide, providing additional functionality to the polypeptide, incorporating a tag, label or detectable signal into the polypeptide, easing the isolation properties of the polypeptide, and any combination of the aforementioned modifications.
  • polypeptides comprising at least one non-natural amino acid or modified non-natural amino acid are further provided and described in U.S. Patent Application Nos. 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 which are.
  • a very wide variety of non-naturally encoded amino acids are suitable for use in the present invention. Any number of non-naturally encoded amino acids can be introduced into a polypeptide.
  • the introduced non-naturally encoded amino acids are substantially chemically inert toward the 20 common, genetically-encoded amino acids (i.e., alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine).
  • amino acids i.e., alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
  • the non-naturally encoded amino acids include side chain functional groups that react efficiently and selectively with functional groups not found in the 20 common amino acids (including but not limited to, azido, ketone, aldehyde and aminooxy groups) to form stable conjugates. Because the non-naturally encoded amino acids of the invention typically differ from the natural amino acids only in the structure of the side chain, the non-naturally encoded amino acids form amide bonds with other amino acids, including but not limited to, natural or non-naturally encoded, in the same manner in which they are formed in naturally occurring polypeptides. However, the non-naturally encoded amino acids have side chain groups that distinguish them from the natural amino acids.
  • the side chain (R group) optionally comprises an alkyl-, aryl-, acyl-, keto-, azido-, hydroxyl-, hydrazine, cyano-, halo-, hydrazide, alkenyl, alkynl, ether, thiol, seleno-, sulfonyl-, borate, boronate, phospho, phosphono, phosphine, heterocyclic, enone, imine, aldehyde, ester, thioacid, hydroxylamine, amino group, or the like or any combination thereof.
  • Non-naturally occurring amino acids of interest include, but are not limited to, amino acids comprising a photoactivatable cross-linker, spin-labeled amino acids, fluorescent amino acids, metal binding amino acids, metal-containing amino acids, radioactive amino acids, amino acids with novel functional groups, amino acids that covalently or noncovalently interact with other molecules, photocaged and/or photoisomerizable amino acids, amino acids comprising biotin or a biotin analogue, glycosylated amino acids such as a sugar substituted serine, other carbohydrate modified amino acids, keto-containing amino acids, amino acids comprising polyethylene glycol or polyether, heavy atom substituted amino acids, chemically cleavable and/or photocleavable amino acids, amino acids with an elongated side chains as compared to natural amino acids, including but not limited to, polyethers or long chain hydrocarbons, including but not limited to, greater than about 5 or greater than about 10 carbons, carbon-linked sugar-containing amino acids,
  • non-natural amino acids for incorporation into polypeptides are found in WO 2002/085923 entitled "In vivo incorporation of unnatural amino acids". Methods and compositions for the in vivo incorporation of non-naturally encoded amino acids are described in U.S. Patent Application Publication 2003/0082575 . Methods for selecting an orthogonal tRNA-tRNA synthetase pair for use in in vivo translation system of an organism are also described in U.S. Patent Application Publications 2003/0082575 and 2003/0108885 . PCT Publication No.
  • WO 04/035743 entitled “Site Specific Incorporation of Keto Amino Acids into Proteins,” describes orthogonal RS and tRNA pairs for the incorporation of keto amino acids.
  • Non-naturally encoded amino acids have side chain groups that distinguish them from the natural amino acids.
  • the side chain may comprises an alkyl-, aryl-, acyl-, keto-, azido-, hydroxyl-, hydrazine, cyano-, halo-, hydrazide, alkenyl, alkynl, ether, thiol, seleno-, sulfonyl-, borate, boronate, phospho, phosphono, phosphine, heterocyclic, enone, imine, aldehyde, ester, thioacid, hydroxylamine, amino group, or the like or any combination thereof.
  • polypeptides with at least one non-natural amino acid or modified non-natural amino acid group include at least one post-translational modification at some position on the polypeptide.
  • the post-translational modification occurs via the cellular machinery (e.g. , glycosylation, acetylation, acylation, lipid-modification, palmitoylation, palmitate addition, phosphorylation, glycolipid-linkage modification, and the like), in many instances, such cellular-machinery-based post-translational modifications occur at the naturally occurring amino acid sites on the polypeptide, however, in certain embodiments, the cellular-machinery-based post-translational modifications occur on the non-natural amino acid site(s) on the polypeptide.
  • the post-translational modification does not utilize the cellular machinery, but is instead providing by attachment of a molecule (including but not limited to, a label; a dye; a polymer; a water-soluble polymer; a derivative of polyethylene glycol; a photocrosslinker; a cytotoxic compound; a drug; an affinity label; a photoaffinity label; a reactive compound; a resin; a second protein or polypeptide or polypeptide analog; an antibody or antibody fragment; a metal chelator; a cofactor; a fatty acid; a carbohydrate; a polynucleotide; a DNA; a RNA; an antisense polynucleotide; a saccharide, a water-soluble dendrimer, a cyclodextrin, an inhibitory ribonucleic acid; a biomaterial; a nanoparticle; a spin label; a fluorophore, a metal-containing moiety; a radioactive moiety
  • the post-translational modification is made in vivo in a eukaryotic cell or in a non-eukaryotic cell. In certain embodiments, the post-translational modification is made in vitro. Also disclosed are methods for producing, purifying, characterizing and using such polypeptides containing at least one such post-translationally modified non-natural amino acids.
  • reagents capable of reacting with a non-natural amino acid that is part of a polypeptide so as to produce any of the aforementioned post-translational modifications are reagents capable of reacting with a non-natural amino acid that is part of a polypeptide so as to produce any of the aforementioned post-translational modifications.
  • the resulting post-translationally modified non-natural amino acid will contain at least one non-natural amino acid which may undergo subsequent modification reactions.
  • methods for producing, purifying, characterizing and using such reagents that are capable of any such post-translational modifications of such non-natural amino acid(s).
  • the protein includes at least one post-translational modification that is made in vivo by one host cell, where the post-translational modification is not normally made by another host cell type. In certain embodiments, the protein includes at least one post-translational modification that is made in vivo by a eukaryotic cell, where the post-translational modification is not normally made by a non-eukaryotic cell. Examples of post-translational modifications include, but are not limited to, glycosylation, acetylation, acylation, lipid-modification, palmitoylation, palmitate addition, phosphorylation, glycolipid-linkage modification, and the like.
  • the post-translational modification comprises attachment of an oligosaccharide to an asparagine by a GlcNAc-asparagine linkage (including but not limited to, where the oligosaccharide comprises (GlcNAc-Man) 2 -Man-GlcNAc-GlcNAc, and the like).
  • the post-translational modification comprises attachment of an oligosaccharide (including but not limited to, Gal-GalNAc, Gal-GlcNAc, etc.
  • a protein or polypeptide can comprise a secretion or localization sequence, an epitope tag, a FLAG tag, a polyhistidine tag, a GST fusion, and/or the like.
  • secretion signal sequences include, but are not limited to, a prokaryotic secretion signal sequence, a eukaryotic secretion signal sequence, a eukaryotic secretion signal sequence 5'-optimized for bacterial expression, a novel secretion signal sequence, pectate lyase secretion signal sequence, Omp A secretion signal sequence, and a phage secretion signal sequence.
  • secretion signal sequences include, but are not limited to, STII (prokaryotic), Fd GIII and M13 (phage), Bg12 (yeast), and the signal sequence bla derived from a transposon. Also disclosed are methods for producing, purifying, characterizing and using such polypeptides containing at least one such post-translational modification.
  • the protein or polypeptide of interest can contain at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or ten or more non-natural amino acids.
  • the non-natural amino acids can be the same or different, for example, there can be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more different sites in the protein that comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more different non-natural amino acids.
  • at least one, but fewer than all, of a particular amino acid present in a naturally occurring version of the protein is substituted with an non-natural amino acid.
  • polypeptides comprising at least one non-natural amino acid.
  • Introduction of at least one non-natural amino acid into a polypeptide can allow for the application of conjugation chemistries that involve specific chemical reactions, including, but not limited to, with one or more non-natural amino acids while not reacting with the commonly occurring 20 amino acids.
  • conjugation chemistries that involve specific chemical reactions, including, but not limited to, with one or more non-natural amino acids while not reacting with the commonly occurring 20 amino acids.
  • the amino acid side chains can then be modified by utilizing chemistry methodologies known to those of ordinary skill in the art to be suitable for the particular functional groups or substituents present in the naturally encoded amino acid.
  • the non-natural amino acid methods and compositions described herein provides conjugates of substances having a wide variety of functional groups, substituents or moieties, with other substances including but not limited to a label; a dye; a polymer; a water-soluble polymer; a derivative of polyethylene glycol; a photocrosslinker; a cytotoxic compound; a drug; an affinity label; a photoaffmity label; a reactive compound; a resin; a second protein or polypeptide or polypeptide analog; an antibody or antibody fragment; a metal chelator; a cofactor; a fatty acid; a carbohydrate; a polynucleotide; a DNA; a RNA; an antisense polynucleotide; a saccharide;, a water-soluble dendrimer; a cyclodextrin; an inhibitory ribonucleic acid; a biomaterial; a nanoparticle; a spin label; a fluorophore, a metal-containing
  • compositions, methods, techniques and strategies further described in U.S. Patent Application Nos. 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 are methods for studying or using any of the aforementioned (modified) non-natural amino acid polypeptides. Included within this aspect, by way of example only, are therapeutic, diagnostic, assay-based, industrial, cosmetic, plant biology, environmental, energy-production, and/or military uses which would benefit from a polypeptide comprising a (modified) non-natural amino acid polypeptides or protein.
  • Non-natural amino acid polypeptides or a fragment thereof can be obtained by combining the non-natural amino acid polypeptides or a fragment thereof with a library of molecules under conditions suitable to allow specific interactions. Also disclosed herein is a method for detecting the aforementioned (modified) non-natural amino acid polypeptides or a fragment thereof where non-natural amino acid polypeptides or a fragment thereof are obtained by combining the non-natural amino acid polypeptides or a fragment thereof with the library of proteins or a portion thereof under conditions suitable to allow specific interaction.
  • Such interactions include but are not limited to acetylation, carboxylation, acylation, phosphorylation, dephosphorylation, ubiquitination, glycosylation, lipid modification, ADP-ribosylation, bioavailability and half-life.
  • Such libraries include alpha-1 antitrypsin, angiostatin, antihemolytic factor, antibody, apolipoprotein, apoprotein, atrial natriuretic factor, atrial natriuretic polypeptide, atrial peptide, C-X-C chemokine, T39765, NAP-2, ENA-78, gro-a, gro-b, gro-c, IP-10, GCP-2, NAP-4, SDF-1, PF4, MIG, calcitonin, c-kit ligand, cytokine, CC chemokine, monocyte chemoattractant protein-1, monocyte chemoattractant protein-2, monocyte chemoattractant protein-3, monocyte inflammatory protein-1 alpha, monocyte inflammatory protein-i beta, RANTES, 1309, R83915, R91733, HCC1, T58847, D31065, T64262, CD40, CD40 ligand, c-kit ligand, collagen, colony stimulating factor
  • the non-natural amino acid polypeptides or a fragment thereof disclosed herein include incorporation of one or more non-natural amino acids into a polypeptide.
  • One or more non-natural amino acids may be incorporated at a particular position which does not disrupt activity of the polypeptide. This can be achieved by making "conservative" substitutions, including but not limited to, substituting hydrophobic amino acids with hydrophobic amino acids, bulky amino acids for bulky amino acids, hydrophilic amino acids for hydrophilic amino acids and/or inserting the non-natural amino acid in a location that is not required for activity.
  • a variety of biochemical and structural approaches can be employed to select the desired sites for substitution with a non-natural amino acid within the polypeptide. Any position of the polypeptide chain is suitable for selection to incorporate a non-natural amino acid, and selection may be based on rational design or by random selection for any or no particular desired purpose.
  • Selection of desired sites may be for producing a non-natural amino acid polypeptide (which may be further modified or remain unmodified) having any desired property or activity, including but not limited to agonists, super-agonists, inverse agonists, antagonists, receptor binding modulators, receptor activity modulators, modulators of binding to one or more binding partners, binding partner activity modulators, binding partner conformation modulators, dimer or multimer formation, no change to activity or property compared to the native molecule, or manipulating any physical or chemical property of the polypeptide such as solubility, aggregation, or stability.
  • locations in the polypeptide required for biological activity of a polypeptide can be identified using point mutation analysis, alanine scanning or homolog scanning methods known in the art.
  • Residues other than those identified as critical to biological activity by alanine or homolog scanning mutagenesis may be good candidates for substitution with a non-natural amino acid depending on the desired activity sought for the polypeptide.
  • the sites identified as critical to biological activity may also be good candidates for substitution with a non-natural amino acid, again depending on the desired activity sought for the polypeptide.
  • Another alternative would be to simply make serial substitutions in each position on the polypeptide chain with a non-natural amino acid and observe the effect on the activities of the polypeptide. It is readily apparent to those of ordinary skill in the art that any means, technique, or method for selecting a position for substitution with a non-natural amino acid into any polypeptide is suitable for use in the present invention.
  • the structure and activity of naturally-occurring mutants of a polypeptide that contain deletions can also be examined to determine regions of the protein that are likely to be tolerant of substitution with a non-natural amino acid. Once residues that are likely to be intolerant to substitution with non-natural amino acids have been eliminated, the impact of proposed substitutions at each of the remaining positions can be examined from the three-dimensional structure of the relevant polypeptide, and any associated ligands or binding proteins. X-ray crystallographic and NMR structures of many polypeptides are available in the Protein Data Bank (PDB, www.rcsb.org), a centralized database containing three-dimensional structural data of large molecules of proteins and nucleic acids. Thus, those of ordinary skill in the art can readily identify amino acid positions that can be substituted with non-natural amino acids.
  • PDB Protein Data Bank
  • Exemplary sites of incorporation of a non-natural amino acid include, but are not limited to, those that are excluded from potential receptor binding regions, regions for binding to one or more binding partners, may be fully or partially solvent exposed, have minimal or no hydrogen-bonding interactions with nearby residues, may be minimally exposed to nearby reactive residues, may be on one or more of the exposed faces of the polypeptide, may be in regions that are highly flexible or structurally rigid as predicted by the three-dimensional, secondary, tertiary, or quaternary structure of the polypeptide, bound or unbound to its associated receptor, ligand or binding proteins., or coupled or not coupled to another polypeptide or other biologically active molecule, or may modulate the conformation of the polypeptide itself or a dimer or multimer comprising one or more polypeptide, by altering the flexibility or rigidity of the complete structure as desired.
  • non-natural amino acids can be substituted for, or incorporated into, a given position in a polypeptide.
  • a particular non-natural amino acid may be selected for incorporation based on an examination of the three dimensional crystal structure of a polypeptide with its associated ligand, receptor and/or binding proteins, secondary, tertiary or quaternary structure, a preference for conservative substitutions (i.e. , aryl-based non-natural amino acids, such as p-acetylphenylalanine or O-propargyltyrosine substituting for Phe, Tyr or Trp), and the specific conjugation chemistry that one desires to introduce into the polypeptide protein.
  • the method further includes incorporating into the protein the non-natural amino acid, where the non-natural amino acid comprises a first reactive group; and contacting the protein with a molecule (including but not limited to a label; a dye; a polymer; a water-soluble polymer; a derivative of polyethylene glycol; a photocrosslinker; a cytotoxic compound; a drug; an affinity label; a photoaffinity label; a reactive compound; a resin; a second protein or polypeptide or polypeptide analog; an antibody or antibody fragment; a metal chelator; a cofactor; a fatty acid; a carbohydrate; a polynucleotide; a DNA; a RNA; an antisense polynucleotide; a saccharide; a water-soluble dendrimer; a cyclodextrin; an inhibitory ribonucleic acid; a biomaterial; a nanoparticle; a spin label; a fluorophore, a metal-
  • the non-natural amino acid substitution(s) or incorporation(s) will be combined with other additions, substitutions, or deletions within the polypeptide to affect other biological traits.
  • the other additions, substitutions or deletions may increase the stability (including but not limited to, resistance to proteolytic degradation) of the polypeptide or increase affinity of the polypeptide for its appropriate receptor, ligand and/or binding proteins.
  • the other additions, substitutions or deletions may increase the solubility (including but not limited to, when expressed in E. coli or other host cells) of the polypeptide.
  • sites are selected for substitution with a naturally encoded or non-natural amino acid in addition to another site for incorporation of a non-natural amino acid for the purpose of increasing the polypeptide solubility following expression in E. coli recombinant host cells.
  • the polypeptides comprise another addition, substitution, or deletion that modulates affinity for the associated ligand, binding proteins, and/or receptor, modulates (including but not limited to, increases or decreases) receptor dimerization, stabilizes receptor dimers, modulates circulating half-life, modulates release or bio-availability, facilitates purification, or improves or alters a particular route of administration.
  • polypeptide can comprise chemical or enzyme cleavage sequences, protease cleavage sequences, reactive groups, antibody-binding domains (including but not limited to, FLAG or poly-His) or other affinity based sequences (including but not limited to, FLAG, poly-His, GST, etc. ) or linked molecules (including but not limited to, biotin) that improve detection (including but not limited to, GFP), purification, transport through tissues or cell membranes, prodrug release or activation, size reduction, or other traits of the polypeptide.
  • antibody-binding domains including but not limited to, FLAG or poly-His
  • affinity based sequences including but not limited to, FLAG, poly-His, GST, etc.
  • linked molecules including but not limited to, biotin
  • the polypeptide can be homologous to a therapeutic protein selected from the group consisting of: alpha-1 antitrypsin, angiostatin, antihemolytic factor, antibody, antibody fragments, apolipoprotein, apoprotein, atrial natriuretic factor, atrial natriuretic polypeptide, atrial peptide, C-X-C chemokine, T39765, NAP-2, ENA-78, gro-a, gro-b, gro-c, IP-10, GCP-2, NAP-4, SDF-1, PF4, MIG, calcitonin, c-kit ligand, cytokine, CC chemokine, monocyte chemoattractant protein-1, monocyte chemoattractant protein-2, monocyte chemoattractant protein-3, monocyte inflammatory protein-1 alpha, monocyte
  • Antibody fragments herein include antibodies that are smaller components that exist within full-length antibodies, and antibodies that have been engineered.
  • Antibody fragments include but are not limited to Fv, Fc, Fab, and (Fab') 2, single chain Fv (scFv), diabodies, triabodies, tetrabodies, bifunctional hybrid antibodies, CDR1, CDR2, CDR3, combinations of CDR's, variable regions, framework regions, constant regions, and the like ( Maynard & Georgiou, 2000, Annu. Rev. Biomed. Eng. 2:339-76 ; Hudson, 1998, Curr. Opin. Biotechnol. 9:395-402 ).
  • Another functional substructure is a single chain Fv (scFv), comprised of the variable regions of the immunoglobulin heavy and light chain, covalently connected by a peptide linker ( S-z Hu et al., 1996, Cancer Research, 56, 3055-3061 ).
  • scFv single chain Fv
  • These small (Mr 25,000) proteins generally retain specificity and affinity for antigen in a single polypeptide and can provide a convenient building block for larger, antigen-specific molecules.
  • Polypeptides also include the antibody heavy chain, light chain, variable region, alternative scaffold non-antibody molecules, and bispecific antibodies, as well as other antigen-binding polypeptides or fragments thereof.
  • GH polypeptides in this application is intended to use the generic term as an example of any member of the GH supergene family.
  • modifications and chemistries described herein with reference to GH polypeptides or protein can be equally applied to any member of the GH supergene family, including those specifically listed herein.
  • the following proteins include those encoded by genes of the growth hormone (GH) supergene family ( Bazan, F., Immunology Today 11: 350-354 (1990 ); Bazan, J. F. Science 257: 410-413 (1992 ); Mott, H. R. and Campbell, I. D., Current Opinion in Structural Biology 5: 114-121 (1995 ); Silvennoinen, O. and Ihle, J.
  • GH growth hormone
  • cytokines including G-CSF ( Zink et al., FEBS Lett. 314:435 (1992 ); Zink et al., Biochemistry 33:8453 (1994 ); Hill et al., Proc. Natl. Acad. Sci. USA 90:5167 (1993 )), GM-CSF ( Diederichs, K., et al. Science 154: 1779-1782 (1991 ); Walter et al., J. Mol. Biol. 224:1075-1085 (1992 )), IL-2 ( Bazan, J. F. and McKay, D. B.
  • G-CSF Zink et al., FEBS Lett. 314:435 (1992 ); Zink et al., Biochemistry 33:8453 (1994 ); Hill et al., Proc. Natl. Acad. Sci. USA 90:5167 (1993 )
  • GM-CSF Diederichs, K., et al. Science
  • EPO is considered to be a member of this family based upon modeling and mutagenesis studies ( Boissel et al., J. Biol. Chem. 268: 15983-15993 (1993 ); Wen et al., J. Biol. Chem. 269: 22839-22846 (1994 )).
  • cytokines and growth factors including ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), thrombopoietin (TPO), oncostatin M, macrophage colony stimulating factor (M-CSF), IL-3, IL-6, IL-7, IL-9, IL-12, IL-13, IL-15, and granulocyte-colony stimulating factor (G-CSF), as well as the IFN's such as alpha, beta, omega, tau, epsilon, and gamma interferon belong to this family (reviewed in Mott and Campbell, Current Opinion in Structural Biology 5: 114-121 (1995 ); Silvennoinen and Ihle (1996) SIGNALLING BY THE HEMATOPOIETIC CYTOKINE RECEPTORS ). All of the above cytokines and growth factors are now considered to comprise one large gene family.
  • CNTF ciliary neurotrophic factor
  • LIF leukemia inhibitory factor
  • TPO
  • GH family members including but not limited to; GH and EPO, bind a single type of receptor and cause it to form homodimers.
  • Other family members including but not limited to, IL-2, IL4. and IL-6, bind more than one type of receptor and cause the receptors to form heterodimers or higher order aggregates ( Davis et al., (1993) Science 260: 1805-1808 ; Paonessa et al., 1995) EMBO J.
  • a general conclusion reached from mutational studies of various members of the GH supergene family is that the loops joining the alpha helices generally tend to not be involved in receptor binding.
  • the short B-C loop appears to be non-essential for receptor binding in most, if not all, family members.
  • the B-C loop may be substituted with non-natural amino acids as described herein in members of the GH supergene family.
  • the A-B loop, the C-D loop (and D-E loop of interferon/ IL-10-like members of the GH superfamily) may also be substituted with a non-natural amino acid.
  • Amino acids proximal to helix A and distal to the final helix also tend not to be involved in receptor binding and also may be sites for introducing non-natural amino acids.
  • a non-natural amino acid is substituted at any position within a loop structure including but not limited to the first 1, 2, 3, 4, 5, 6, 7, or more amino acids of the A-B, B-C, C-D or D-E loop.
  • a non-natural amino acid is substituted within the last 1, 2, 3, 4, 5, 6, 7, or more amino acids of the A-B, B-C, C-D or D-E loop.
  • Certain members of the GH family including but not limited to, EPO, IL-2, IL-3, IL-4, IL-6, IFN, GM-CSF, TPO, IL-10, IL-12 p35, IL-13, IL-15 and beta interferon contain N-linked and/or O-linked sugars.
  • the glycosylation sites in the proteins occur almost exclusively in the loop regions and not in the alpha helical bundles. Because the loop regions generally are not involved in receptor binding and because they are sites for the covalent attachment of sugar groups, they may be useful sites for introducing non-natural amino acid substitutions into the proteins.
  • Amino acids that comprise the N- and O-linked glycosylation sites in the proteins may be sites for non-natural amino acid substitutions because these amino acids are surface-exposed. Therefore, the natural protein can tolerate bulky sugar groups attached to the proteins at these sites and the glycosylation sites tend to be located away from the receptor binding sites.
  • Additional members of the GH gene family are likely to be discovered in the future. New members of the GH supergene family can be identified through computer-aided secondary and tertiary structure analyses of the predicted protein sequences. Members of the GH supergene family typically possess four or five amphipathic helices joined by non-helical amino acids (the loop regions). The proteins may contain a hydrophobic signal sequence at their N-terminus to promote secretion from the cell. Such later discovered members of the GH supergene family also are included within the methods and compositions described herein. International Patent Application entitled "Modified Four Helical Bundle Polypeptides and Their Uses" ( WO 05/074650 on August 18, 2005 ), provides methods for site selection and incorporation of non-natural amino acids into polypeptides.
  • non-natural amino acids are suitable for use in the methods and compositions described herein as long as the non-natural amino acid has at least one of the following four properties: (1) at least one functional group on the sidechain of the non-natural amino acid with at least one characteristic and/or activity and/or reactivity orthogonal to the chemical reactivity of the 20 common, genetically-encoded amino acids (i.e., alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine), or at least orthogonal to the chemical reactivity of the naturally occurring amino acids present in the polypeptide that includes the non-natural amino acid; (2) the introduced non-natural amino acid is substantially chemically inert toward the 20 common, genetically-en
  • Non-natural amino acids that satisfy these four properties for non-natural amino acids that can be used with the compositions and methods further described in U.S. Patent Application Nos. 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 . Any number of non-natural amino acids can be introduced into the polypeptide. Non-natural amino acids may also include protected or masked oximes or protected or masked groups that can be transformed into an oxime group after deprotection of the protected group or unmasking of the masked group.
  • Non-natural amino acids of interest that may be suitable for use in the methods and compositions described herein include, but are not limited to, amino acids comprising a photoactivatable cross-linker, spin-labeled amino acids, fluorescent amino acids, metal binding amino acids, metal-containing amino acids, radioactive amino acids, amino acids with novel functional groups, amino acids that covalently or noncovalently interact with other molecules, photocaged and/or photoisomerizable amino acids, amino acids comprising biotin or a biotin analogue, glycosylated amino acids such as a sugar substituted serine, other carbohydrate modified amino acids, keto-containing amino acids, amino acids comprising polyethylene glycol or polyether, heavy atom substituted amino acids, chemically cleavable and/or photocleavable amino acids, amino acids with an elongated side chains as compared to natural amino acids, including but not limited to, polyethers or long chain hydrocarbons, including but not limited to, greater than about 5 or greater than about 10 carbons, carbon-linked sugar-containing
  • non-natural amino acids comprise a saccharide moiety.
  • examples of such amino acids include N -acetyl-L-glucosaminyl-L-serine, N -acetyl-L-galactosaminyl-L-serine, N -acetyl-L-glucosaminyl-L-threonine, N -acetyl-L-glucosaminyl-L-asparagine and O -mannosaminyl-L-serine.
  • amino acids also include examples where the naturally-occurring N- or O- linkage between the amino acid and the saccharide is replaced by a covalent linkage not commonly found in nature - including but not limited to, an alkene, an oxime, a thioether, an amide and the like.
  • amino acids also include saccharides that are not commonly found in naturally-occurring proteins such as 2-deoxy-glucose, 2-deoxygalactose and the like.
  • the chemical moieties via non-natural amino acids that can be incorporated into proteins offer a variety of advantages and manipulations of the protein.
  • the unique reactivity of a carbonyl functional group (including a keto functional group) allows selective modification of proteins with any of a number of hydrazine- or hydroxylamine-containing reagents in vitro and in vivo.
  • a heavy atom non-natural amino acid for example, can be useful for phasing X-ray structure data.
  • the site-specific introduction of heavy atoms using non-natural amino acids also provides selectivity and flexibility in choosing positions for heavy atoms.
  • Photoreactive non-natural amino acids include but not limited to, amino acids with benzophenone and arylazides (including but not limited to, phenylazide) side chains), for example, allow for efficient in vivo and in vitro photocrosslinking of protein.
  • photoreactive non-natural amino acids include, but are not limited to, p-azido-phenylalanine and p-benzoyl-phenylalanine.
  • the protein with the photoreactive non-natural amino acids can then be crosslinked at will by excitation of the photoreactive group-providing temporal control.
  • the methyl group of a non-natural amino can be substituted with an isotopically labeled, including but not limited to, methyl group, as a probe of local structure and dynamics, including but not limited to, with the use of nuclear magnetic resonance and vibrational spectroscopy.
  • non-naturally encoded amino acids are commercially available, e.g., from Sigma-Aldrich (St. Louis, MO, USA), Novabiochem (a division of EMD Biosciences, Darmstadt, Germany), or Peptech (Burlington, MA, USA). Those that are not commercially available are optionally synthesized.
  • Organic synthesis techniques see, e.g., Organic Chemistry by Fessendon and Fessendon, (1982, Second Edition, Willard Grant Press, Boston Mass .); Advanced Organic Chemistry by March (Third Edition, 1985, Wiley and Sons, New York ); and Advanced Organic Chemistry by Carey and Sundberg (Third Edition, Parts A and B, 1990, Plenum Press, New York ).
  • Many non-natural amino acids are based on natural amino acids, such as tyrosine, glutamine, phenylalanine, and the like.
  • Non-natural amino acid uptake by a eukaryotic cell is one issue that is typically considered when designing and selecting non-natural amino acids, including but not limited to, for incorporation into a protein.
  • the high charge density of ⁇ -amino acids suggests that these compounds are unlikely to be cell permeable.
  • Natural amino acids are taken up into the eukaryotic cell via a collection of protein-based transport systems. A rapid screen can be done which assesses which non-natural amino acids, if any, are taken up by cells. See, e.g., the toxicity assays in, e.g., U.S. Patent Publication No. US 2004/0198637 entitled "Protein Arrays,"; and Liu, D.R.
  • biosynthetic pathways already exist in cells for the production of amino acids and other compounds. While a biosynthetic method for a particular non-natural amino acid may not exist in nature, including but not limited to, in a eukaryotic cell, the methods and compositions described herein include such methods.
  • biosynthetic pathways for non-natural amino acids are optionally generated in host cell by adding new enzymes or modifying existing host cell pathways. Additional new enzymes are optionally naturally occurring enzymes or artificially evolved enzymes.
  • the biosynthesis of p- aminophenylalanine (as presented in an example in WO 2002/085923 entitled "In vivo incorporation of unnatural amino acids") relies on the addition of a combination of known enzymes from other organisms.
  • the genes for these enzymes can be introduced into a eukaryotic cell by transforming the cell with a plasmid comprising the genes.
  • the genes when expressed in the cell, provide an enzymatic pathway to synthesize the desired compound. Examples of the types of enzymes that are optionally added are provided in the examples below. Additional enzymes sequences are found, for example, in Genbank. Artificially evolved enzymes are also optionally added into a cell in the same manner. In this manner, the cellular machinery and resources of a cell are manipulated to produce non-natural amino acids.
  • recursive recombination including but not limited to, as developed by Maxygen, Inc. (available on the World Wide Web at www.maxygen.com), is optionally used to develop novel enzymes and pathways. See, e.g., Stemmer (1994), Rapid evolution of a protein in vitro by DNA shuffling, Nature 370(4):389-391 ; and, Stemmer, (1994), DNA shuffling by random fragmentation and reassembly: In vitro recombination for molecular evolution, Proc. Natl. Acad. Sci. USA., 91:10747-10751 .
  • DesignPathTM developed by Genencor (available on the World Wide Web at genencor.com) is optionally used for metabolic pathway engineering, including but not limited to, to engineer a pathway to create O-methyl-L-tyrosine in a cell.
  • This technology reconstructs existing pathways in host organisms using a combination of new genes, including but not limited to, identified through functional genomics, and molecular evolution and design.
  • Diversa Corporation (available on the world wide web at diversa.com) also provides technology for rapidly screening libraries of genes and gene pathways, including but not limited to, to create new pathways.
  • the non-natural amino acid produced with an engineered biosynthetic pathway is produced in a concentration sufficient for efficient protein biosynthesis, including but not limited to, a natural cellular amount, but not to such a degree as to affect the concentration of the other amino acids or exhaust cellular resources.
  • concentrations produced in vivo in this manner are about 10 mM to about 0.05 mM.
  • the non-natural amino acid may be present at any location on the polypeptide, including any terminal position or any internal position of the polypeptide.
  • the non-natural amino acid polypeptides described herein may be produced biosynthetically or non-biosyntheticially.
  • biosynthetically is meant any method utilizing a translation system (cellular or non-cellular), including use of at least one of the following components: a polynucleotide, a codon, a tRNA, and a ribosome.
  • non-biosynthetically any method not utilizing a translation system: this approach can be further divided into methods utilizing solid state peptide synthetic methods, solid phase peptide synthetic methods, methods that utilize at least one enzyme, and methods that do not utilize at least one enzyme; of course any of this sub-divisions may overlap and many methods may utilize a combination of these sub-divisions.
  • polypeptides may include but are not limited to at least one non-natural amino acids further described in U.S. Patent Application Nos. 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 ; U.S. Patent Application Publications 2003/0082575 and 2003/0108885 , WO 04/035743 entitled "Site Specific Incorporation of Keto Amino Acids into Proteins," PCT Publication No.
  • WO 04/094593 entitled “Expanding the Eukaryotic Genetic Code”
  • PCT Publication WO 05/074650 entitled “Modified Four Helical Bundle Polypeptides and Their Uses,”.
  • the non-natural amino acid polypeptides may be further modified as described in U.S. Patent Application Nos. 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 ; U.S. Patent Application Publications 2003/0082575 and 2003/0108885 , WO 04/035743 entitled "Site Specific Incorporation of Keto Amino Acids into Proteins," PCT Publication No.
  • a composition includes at least one protein with at least one, including but not limited to, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten or more non-natural amino acids.
  • the polypeptides may comprise one or more natural amino acid substitutions.
  • non-natural amino acid polypeptides further described in U.S. Patent Application Nos. 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 may be chemically synthesized via solid phase peptide synthesis methods (e.g., on a solid resin), by solution phase peptide synthesis methods, and/or without the aid of enzymes
  • other embodiments of the non-natural amino acid polypeptides described herein allow synthesis via a cell membrane, cellular extract, or lysate system or via an in vivo system, i.e., using the cellular machinery of a prokarote or eukaryote cell.
  • a nucleotide sequence encoding a polypeptide comprising a non-natural amino acid may be synthesized on the basis of the amino acid sequence of the parent polypeptide, and then changing the nucleotide sequence so as to effect introduction ( i.e., incorporation or substitution) or removal ( i.e., deletion or substitution) of the relevant amino acid residue(s).
  • the nucleotide sequence may be conveniently modified by site-directed mutagenesis in accordance with conventional methods.
  • the nucleotide sequence may be prepared by chemical synthesis, including but not limited to, by using an oligonucleotide synthesizer, wherein oligonucleotides are designed based on the amino acid sequence of the desired polypeptide, and preferably selecting those codons that are favored in the host cell in which the recombinant polypeptide will be produced.
  • oligonucleotides are designed based on the amino acid sequence of the desired polypeptide, and preferably selecting those codons that are favored in the host cell in which the recombinant polypeptide will be produced.
  • several small oligonucleotides coding for portions of the desired polypeptide may be synthesized and assembled by PCR, ligation or ligation chain reaction. See, e.g., Barany, et al., Proc. Natl. Acad. Sci. 88: 189-193 (1991 ); U.S. 6,521,427 .
  • a selector codon includes, but is not limited to, a unique three base codon, a nonsense codon, such as a stop codon, including but not limited to, an amber codon (UAG), or an opal codon (UGA), an ochre codon, a unnatural codon, a four or more base codon, a rare codon, or the like.
  • a selector codon includes, but is not limited to, a unique three base codon, a nonsense codon, such as a stop codon, including but not limited to, an amber codon (UAG), or an opal codon (UGA), an ochre codon, a unnatural codon, a four or more base codon, a rare codon, or the like.
  • There is a wide range in the number of selector codons that can be introduced into a desired gene including but not limited to, one or more, two or more, three or more, 4, 5, 6, 7, 8, 9, 10 or more in a single polynu
  • selector codons that is a stop codon for the incorporation of one or more non-natural amino acids in vivo.
  • the incorporation of non-natural amino acids in vivo can be done without significant perturbation of the eukaryotic host cell.
  • Selector codons also comprise extended codons, including but not limited to, four or more base codons, such as, four, five, six or more base codons.
  • a selector codon can also include one of the natural three base codons, where the endogenous system does not use (or rarely uses) the natural base codon.
  • Selector codons optionally include unnatural base pairs. These unnatural base pairs further expand the existing genetic alphabet.
  • the unnatural nucleoside is membrane permeable and is phosphorylated to form the corresponding triphosphate.
  • the increased genetic information is stable and not destroyed by cellular enzymes.
  • a translational bypassing system can also be used to incorporate a non-natural amino acid in a desired polypeptide.
  • the protein or polypeptide of interest (or portion thereof) is encoded by a nucleic acid.
  • the nucleic acid comprises at least one selector codon, at least two selector codons, at least three selector codons, at least four selector codons, at least five selector codons, at least six selector codons, at least seven selector codons, at least eight selector codons, at least nine selector codons, ten or more selector codons.
  • the polypeptides can be generated in vivo using modified tRNA and tRNA synthetases to add to or substitute amino acids that are not encoded in naturally-occurring systems. All the methods for generating, screening methods and organisms used for in vivo generation of polypeptides comprising non-natural amino acids which are further described in U.S. Patent Application Nos. 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 ; U.S. Patent Application Publications 2003/0082575 and 2003/0108885 , PCT Publication No. WO 04/094593 entitled "Expanding the Eukaryotic Genetic Code," and PCT Publication WO 05/074650 entitled "Modified Four Helical Bundle Polypeptides and Their Uses,”.
  • tRNAs and tRNA synthetases which use amino acids that are not encoded in naturally-occurring systems are described in, e.g., U.S. Patent Application Publications 2003/0082575 and 2003/0108885 . These methods involve generating a translational machinery that functions independently of the synthetases and tRNAs endogenous to the translation system (and are therefore sometimes referred to as "orthogonal").
  • the translation system comprises an orthogonal tRNA (O-tRNA) and an orthogonal aminoacyl tRNA synthetase (O-RS).
  • orthogonal tRNAs and aminoacyl tRNA synthetases have been described in the art for inserting particular synthetic amino acids into polypeptides, and are generally suitable for in the methods to produce the non-natural amino acid polypeptides.
  • O-tRNA/aminoacyl-tRNA synthetases involves selection of a specific codon which encodes the non-natural amino acid. While any codon can be used, it is generally desirable to select a codon that is rarely or never used in the cell in which the O-tRNA/aminoacyl-tRNA synthetase is expressed. Specific selector codon(s) can be introduced into appropriate positions in the polynucleotide coding sequence using mutagenesis methods known in the art (including but not limited to, site-specific mutagenesis, cassette mutagenesis, restriction selection mutagenesis, etc. ).
  • Suitable bacterial promoters are well known in the art and described, e.g., in Sambrook et al. and Ausubel et al. Bacterial expression systems and eukaryotic host cell or non-eukaryotic host cell systems further described in in U.S. Patent Application Nos.
  • U.S. Patent Application Publications 2003/0082575 and 2003/0108885 PCT Publication No. WO 04/094593 entitled “Expanding the Eukaryotic Genetic Code”
  • PCT Publication WO 05/074650 entitled “Modified Four Helical Bundle Polypeptides and Their Uses” may be used to biosynthesize proteins that comprise non-natural amino acids in large useful quantities.
  • the desired polypeptide may be expressed in any number of suitable expression systems including, for example, yeast, insect cells, mammalian cells, Pseudomonas cells, and bacteria.
  • suitable expression systems including, for example, yeast, insect cells, mammalian cells, Pseudomonas cells, and bacteria.
  • a description of exemplary expression systems is further described in U.S. Patent Application Nos. 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 ; U.S. Patent Application Publications 2003/0082575 and 2003/0108885 , PCT Publication No. WO 04/094593 entitled "Expanding the Eukaryotic Genetic Code," and PCT Publication WO 05/074650 entitled "Modified Four Helical Bundle Polypeptides and Their Uses,"
  • isolation steps Any one of a variety of isolation steps may be performed on the cell lysate, extract, culture medium, inclusion bodies, periplasmic space of the host cells, cytoplasm of the host cells, or other material comprising the desired polypeptide or mixtures resulting from any isolation steps including, but not limited to, affinity chromatography, ion exchange chromatography, hydrophobic interaction chromatography, gel filtration chromatography, high performance liquid chromatography ("HPLC”), reversed phase-HPLC (“RP-HPLC”), expanded bed adsorption, or any combination and/or repetition thereof and in any appropriate order.
  • HPLC high performance liquid chromatography
  • RP-HPLC reversed phase-HPLC
  • expanded bed adsorption or any combination and/or repetition thereof and in any appropriate order.
  • proteins or polypeptides of interest By producing proteins or polypeptides of interest with at least one non-natural amino acid in eukaryotic cells, proteins or polypeptides include eukaryotic post-translational modifications.
  • a protein includes at least one non-natural amino acid and at least one post-translational modification that is made in vivo by a eukaryotic cell, where the post-translational modification is not made by a prokaryotic cell.
  • the post-translation modification is further described in U.S. Patent Application Nos.
  • non-natural amino acid presents additional chemical moieties that can be used to add additional molecules. These modifications can be made in vivo in a eukaryotic or non-eukaryotic cell, or in vitro. Thus, in certain embodiments, the post-translational modification is through the non-natural amino acid.
  • Induction of expression of the recombinant protein results in the accumulation of a protein containing the unnatural analog.
  • o, m and p-fluorophenylalanines have been incorporated into proteins, and exhibit two characteristic shoulders in the UV spectrum which can be easily identified, see, e.g., C. Minks, R. Huber, L. Moroder and N. Budisa, Anal. Biochem., 284:29 (2000 ); trifluoromethionine has been used to replace methionine in bacteriophage T4 lysozyme to study its interaction with chitooligosaccharide ligands by 19 F NMR, see, e.g., H. Duewel, E.
  • Methionine analogs with alkene or alkyne functionalities have also been incorporated efficiently, allowing for additional modification of proteins by chemical means. See, e.g., J. C. van Hest and D. A. Tirrell, FEBS Lett., 428:68 (1998 ); J. C.. van Hest, K. L. Kiick and D. A. Tirrell, J. Am. Chem. Soc., 122:1282 (2000 ); and, K. L. Kiick and D. A. Tirrell, Tetrahedron, 56:9487 (2000 ); U.S. Patent No. 6,586,207 ; U.S. Patent Publication 2002/0042097 .
  • ValRS can misaminoacylate tRNAVal with Cys, Thr, or aminobutyrate (Abu); these noncognate amino acids are subsequently hydrolyzed by the editing domain.
  • a mutant Escherichia coli strain was selected that has a mutation in the editing site of ValRS. This edit-defective ValRS incorrectly charges tRNAVal with Cys.
  • Abu sterically resembles Cys (-SH group of Cys is replaced with -CH3 in Abu)
  • the mutant ValRS also incorporates Abu into proteins when this mutant Escherichia coli strain is grown in the presence of Abu. Mass spectrometric analysis shows that about 24% of valines are replaced by Abu at each valine position in the native protein.
  • Chemical modification has been used to introduce a variety of unnatural side chains, including cofactors, spin labels and oligonucleotides into proteins in vitro. See, e.g., Corey, D.R., Schultz, P.G. Generation of a hybrid sequence-specific single-stranded deoxyribonuclease, Science, 238(4832):1401-1403 (1987 ); Kaiser, E.T., Lawrence D.S., Rokita, S.E. The chemical modification of enzymatic specificity, Annu Rev Biochem, 54:565-595 (1985 ); Kaiser, E.T., Lawrence, D.S.
  • biosynthetic methods that employ chemically modified aminoacyl-tRNAs have been used to incorporate several biophysical probes into proteins synthesized in vitro. See the following publications and references cited within: Brunner, J. New Photolabeling and crosslinking methods, Annu. Rev Biochem, 62:483-514 (1993 ); and, Krieg, U.C., Walter, P., Hohnson, A.E. Photocrosslinking of the signal sequence of nascent preprolactin of the 54-kilodalton polypeptide of the signal recognition particle, Proc. Natl. Acad. Sci, 83(22):8604-8608 (1986 ).
  • unnatural amino acids can be site-specifically incorporated into proteins in vitro by the addition of chemically aminoacylated suppressor tRNAs to protein synthesis reactions programmed with a gene containing a desired amber nonsense mutation.
  • chemically aminoacylated suppressor tRNAs to protein synthesis reactions programmed with a gene containing a desired amber nonsense mutation.
  • close structural homologues e.g., fluorophenylalanine for phenylalanine
  • strains auxotropic for a particular amino acid.
  • a suppressor tRNA was prepared that recognized the stop codon UAG and was chemically aminoacylated with an unnatural amino acid.
  • Conventional site-directed mutagenesis was used to introduce the stop codon TAG, at the site of interest in the protein gene. See, e.g., Sayers, J.R., Schmidt, W. Eckstein, F. 5'-3' Exonucleases in phosphorothioate-based olignoucleotide-directed mutagensis, Nucleic Acids Res, 16(3):791-802 (1988 ).
  • a tRNA may be aminoacylated with a desired amino acid by any method or technique, including but not limited to, chemical or enzymatic aminoacylation.
  • Aminoacylation may be accomplished by aminoacyl tRNA synthetases or by other enzymatic molecules, including but not limited to, ribozymes.
  • ribozyme is interchangeable with "catalytic RNA.”
  • Cech and coworkers Cech, 1987, Science, 236:1532-1539 ; McCorkle et al., 1987, Concepts Biochem. 64:221-226 ) demonstrated the presence of naturally occurring RNAs that can act as catalysts (ribozymes).
  • these natural RNA catalysts have only been shown to act on ribonucleic acid substrates for cleavage and splicing, the recent development of artificial evolution of ribozymes has expanded the repertoire of catalysis to various chemical reactions.
  • RNA molecules that can catalyze aminoacyl-RNA bonds on their own (2')3'-termini ( Illangakekare et al., 1995 Science 267:643-647 ), and an RNA molecule which can transfer an amino acid from one RNA molecule to another ( Lohse et al., 1996, Nature 381:442-444 ).
  • U.S. Patent Application Publication 2003/0228593 describes methods to construct ribozymes and their use in aminoacylation of tRNAs with naturally encoded and non-naturally encoded amino acids.
  • Substrate-immobilized forms of enzymatic molecules that can aminoacylate tRNAs may enable efficient affinity purification of the aminoacylated products.
  • suitable substrates include agarose, sepharose, and magnetic beads. The production and use of a substrate-immobilized form of ribozyme for aminoacylation is described in Chemistry and Biology 2003, 10:1077-1084 and U.S. Patent Application Publication 2003/0228593 .
  • Chemical aminoacylation methods include, but are not limited to, those introduced by Hecht and coworkers ( Hecht, S. M. Acc. Chem. Res. 1992, 25, 545 ; Heckler, T. G.; Roesser, J. R.; Xu, C.; Chang, P.; Hecht, S. M. Biochemistry 1988, 27, 7254 ; Hecht, S. M.; Alford, B. L.; Kuroda, Y.; Kitano, S. J. Biol. Chem. 1978, 253, 4517 ) and by Schultz, Chamberlin, Dougherty and others ( Cornish, V. W.; Mendel, D.; Schultz, P. G. Angew. Chem. Int. Ed. Engl.
  • Methods for generating catalytic RNA may involve generating separate pools of randomized ribozyme sequences, performing directed evolution on the pools, screening the pools for desirable aminoacylation activity, and selecting sequences of those ribozymes exhibiting desired aminoacylation activity.
  • Ribozymes can comprise motifs and/or regions that facilitate acylation activity, such as a GGU motif and a U-rich region.
  • a GGU motif can facilitate recognition of an amino acid substrate
  • a GGU-motif can form base pairs with the 3' termini of a tRNA.
  • the GGU and motif and U-rich region facilitate simultaneous recognition of both the amino acid and tRNA simultaneously, and thereby facilitate aminoacylation of the 3' terminus of the tRNA.
  • Ribozymes can be generated by in vitro selection using a partially randomized r24mini conjugated with tRNA Asn CCCG, followed by systematic engineering of a consensus sequence found in the active clones.
  • An exemplary ribozyme obtained by this method is termed "Fx3 ribozyme" and is described in U.S. Pub. App. No. 2003/0228593 , acts as a versatile catalyst for the synthesis of various aminoacyl-tRNAs charged with cognate non-natural amino acids.
  • Aminoacylate tRNAs ribozymes can be immobilized on a substrate so as to enable efficient affinity purification of the aminoacylated tRNAs.
  • suitable substrates include, but are not limited to, agarose, sepharose, and magnetic beads.
  • Ribozymes can be immobilized on resins by taking advantage of the chemical structure of RNA, such as the 3'-cis-diol on the ribose of RNA can be oxidized with periodate to yield the corresponding dialdehyde to facilitate immobilization of the RNA on the resin.
  • Various types of resins can be used including inexpensive hydrazide resins wherein reductive amination makes the interaction between the resin and the ribozyme an irreversible linkage. Synthesis of aminoacyl-tRNAs can be significantly facilitated by this on-column aminoacylation technique. Kourouklis et al. Methods 2005; 36:239-4 describe a column-based aminoacylation system.
  • Isolation of the aminoacylated tRNAs can be accomplished in a variety of ways.
  • One suitable method is to elute the aminoacylated tRNAs from a column with a buffer such as a sodium acetate solution with 10 mM EDTA, a buffer containing 50 mM N-(2-hydroxyethyl)piperazine-N'-(3-propanesulfonic acid), 12.5 mM KCl, pH 7.0, 10 mM EDTA, or simply an EDTA buffered water (pH 7.0).
  • a buffer such as a sodium acetate solution with 10 mM EDTA, a buffer containing 50 mM N-(2-hydroxyethyl)piperazine-N'-(3-propanesulfonic acid), 12.5 mM KCl, pH 7.0, 10 mM EDTA, or simply an EDTA buffered water (pH 7.0).
  • the aminoacylated tRNAs can be added to translation reactions in order to incorporate the amino acid with which the tRNA was aminoacylated in a position of choice in a polypeptide made by the translation reaction.
  • Examples of translation systems in which the aminoacylated tRNAs of the present invention may be used include, but are not limited to cell lysates. Cell lysates provide reaction components necessary for in vitro translation of a polypeptide from an input mRNA. Examples of such reaction components include but are not limited to ribosomal proteins, rRNA, amino acids, tRNAs, GTP, ATP, translation initiation and elongation factors and additional factors associated with translation. Additionally, translation systems may be batch translations or compartmentalized translation. Batch translation systems combine reaction components in a single compartment while compartmentalized translation systems separate the translation reaction components from reaction products that can inhibit the translation efficiency. Such translation systems are available commercially.
  • Coupled transcription/translation systems allow for both transcription of an input DNA into a corresponding mRNA, which is in turn translated by the reaction components.
  • An example of a commercially available coupled transcription/translation is the Rapid Translation System (RTS, Roche Inc.).
  • the system includes a mixture containing E. coli lysate for providing translational components such as ribosomes and translation factors.
  • an RNA polymerase is included for the transcription of the input DNA into an mRNA template for use in translation.
  • RTS can use compartmentalization of the reaction components by way of a membrane interposed between reaction compartments, including a supply/waste compartment and a transcription/translation compartment.
  • Aminoacylation of tRNA may be performed by other agents, including but not limited to, transferases, polymerases, catalytic antibodies, multi-functional proteins, and the like.
  • Microinjection techniques have also been use incorporate unnatural amino acids into proteins. See , e.g ., M. W. Nowak, P. C. Kearney, J. R. Sampson, M. E. Saks, C. G. Labarca, S. K. Silverman, W. G. Zhong, J. Thorson, J. N. Abelson, N. Davidson, P. G. Schultz, D. A. Dougherty and H. A. Lester, Science, 268:439 (1995 ); and, D. A. Dougherty, Curr. Opin. Chem. Biol., 4:645 (2000 ).
  • a Xenopus oocyte was coinjected with two RNA species made in vitro: an mRNA encoding the target protein with a UAG stop codon at the amino acid position of interest and an amber suppressor tRNA aminoacylated with the desired unnatural amino acid.
  • the translational machinery of the oocyte then inserts the unnatural amino acid at the position specified by UAG.
  • This method has allowed in vivo structure-function studies of integral membrane proteins, which are generally not amenable to in vitro expression systems. Examples include the incorporation of a fluorescent amino acid into tachykinin neurokinin-2 receptor to measure distances by fluorescence resonance energy transfer, see, e.g., G. Turcatti, K. Nemeth, M. D. Edgerton, U.
  • the ability to incorporate unnatural amino acids directly into proteins in vivo offers the advantages of high yields of mutant proteins, technical ease, the potential to study the mutant proteins in cells or possibly in living organisms and the use of these mutant proteins in therapeutic treatments.
  • the ability to include unnatural amino acids with various sizes, acidities, nucleophilicities, hydrophobicities, and other properties into proteins can greatly expand our ability to rationally and systematically manipulate the structures of proteins, both to probe protein function and create new proteins or organisms with novel properties.
  • the process is difficult, because the complex nature of tRNA-synthetase interactions that are required to achieve a high degree of fidelity in protein translation.
  • a yeast amber suppressor tRNAPheCUA /phenylalanyl-tRNA synthetase pair was used in a p-F-Phe resistant, Phe auxotrophic Escherichia coli strain. See, e.g., R. Furter, Protein Sci., 7:419 (1998 ). It may also be possible to obtain expression of a polynucleotide using a cell-free (in-vitro) translational system. Translation systems may be cellular or cell-free, and may be prokaryotic or eukaryotic.
  • Cellular translation systems include, but are not limited to, whole cell preparations such as permeabilized cells or cell cultures wherein a desired nucleic acid sequence can be transcribed to mRNA and the mRNA translated.
  • Cell-free translation systems are commercially available and many different types and systems are well-known. Examples of cell-free systems include, but are not limited to, prokaryotic lysates such as Escherichia coli lysates, and eukaryotic lysates such as wheat germ extracts, insect cell lysates, rabbit reticulocyte lysates, rabbit oocyte lysates and human cell lysates.
  • Eukaryotic extracts or lysates may be preferred when the resulting protein is glycosylated, phosphorylated or otherwise modified because many such modifications are only possible in eukaryotic systems.
  • Some of these extracts and lysates are available commercially (Promega; Madison, Wis.; Stratagene; La Jolla, Calif.; Amersham; Arlington Heights, Ill.; GIBCO/BRL; Grand Island, N.Y.).
  • Membranous extracts such as the canine pancreatic extracts containing microsomal membranes, are also available which are useful for translating secretory proteins.
  • Reconstituted translation systems may also be used. Mixtures of purified translation factors have also been used successfully to translate mRNA into protein as well as combinations of lysates or lysates supplemented with purified translation factors such as initiation factor-1 (IF-1), IF-2, IF-3 ( ⁇ or ⁇ ), elongation factor T (EF-Tu), or termination factors. Cell-free systems may also be coupled transcription/translation systems wherein DNA is introduced to the system, transcribed into mRNA and the mRNA translated as described in Current Protocols in Molecular Biology (F. M. Ausubel et al. editors, Wiley Interscience, 1993 ).
  • RNA transcribed in eukaryotic transcription system may be in the form of heteronuclear RNA (hnRNA) or 5'-end caps (7-methyl guanosine) and 3'-end poly A tailed mature mRNA, which can be an advantage in certain translation systems.
  • hnRNA heteronuclear RNA
  • 5'-end caps (7-methyl guanosine) and 3'-end poly A tailed mature mRNA which can be an advantage in certain translation systems.
  • capped mRNAs are translated with high efficiency in the reticulocyte lysate system.
  • non-natural amino acid polypeptides described above are useful for, including but not limited to, novel therapeutics, diagnostics, catalytic enzymes, industrial enzymes, binding proteins and including but not limited to, the study of protein structure and function. See, e.g., Dougherty, (2000) Unnatural Amino Acids as Probes of Protein Structure and Function, Current Opinion in Chemical Biology, 4:645-652 .
  • Other uses for the non-natural amino acid polypeptides described above include, by way of example only, assay-based, cosmetic, plant biology, environmental, energy-production, and/or military uses.
  • non-natural amino acid polypeptides described above can undergo further modifications so as to incorporate new or modified functionalities, including manipulating the therapeutic effectiveness of the polypeptide, improving the safety profile of the polypeptide, adjusting the pharmacokinetics, pharmacologics and/or pharmacodynamics of the polypeptide (e . g ., increasing water solubility, bioavailability, increasing serum half-life, increasing therapeutic half-life, modulating immunogenicity, modulating biological activity, or extending the circulation time), providing additional functionality to the polypeptide, incorporating a tag, label or detectable signal into the polypeptide, easing the isolation properties of the polypeptide, and any combination of the aforementioned modifications.
  • new or modified functionalities including manipulating the therapeutic effectiveness of the polypeptide, improving the safety profile of the polypeptide, adjusting the pharmacokinetics, pharmacologics and/or pharmacodynamics of the polypeptide (e . g ., increasing water solubility, bioavailability, increasing serum half-life, increasing
  • compositions, strategies and techniques described herein are not limited to a particular type, class or family of polypeptides or proteins. Indeed, virtually any polypeptides may include at least one non-natural amino acid.
  • a composition may include at least one protein with at least one, including but not limited to, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten or more non-natural amino acids that have been post-translationally modified.
  • the post-translationally-modified non-natural amino acids can be the same or different, including but not limited to, there can be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more different sites in the protein that comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more different post-translationally-modified non-natural amino acids.
  • a composition may include a protein with at least one, but fewer than all, of a particular amino acid present in the protein as substituted with the post-translationally-modified non-natural amino acid.
  • the post-translationally-modified non-natural amino acids can be identical or different (including but not limited to, the protein can include two or more different types of post-translationally-modified non-natural amino acids, or can include two of the same post-translationally-modified non-natural amino acid).
  • the post-translationally-modified non-natural amino acids can be the same, different or a combination of a multiple post-translationally-modified non-natural amino acid of the same kind with at least one different post-translationally-modified non-natural amino acid.
  • the post-translational modification can be through a nucleophilic-electrophilic reaction.
  • Most reactions currently used for the selective modification of proteins involve covalent bond formation between nucleophilic and electrophilic reaction partners, including but not limited to the reaction of ⁇ -haloketones with histidine or cysteine side chains. Selectivity in these cases is determined by the number and accessibility of the nucleophilic residues in the protein.
  • other more selective reactions can be used such as the reaction of an unnatural keto-amino acid with hydrazides or aminooxy compounds, in vitro and in vivo. See, e.g ., Cornish, et al., (1996) J. Am. Chem.
  • non-natural amino acid components which includes non-natural amino acids, as well as the non-natural amino acid portion of a polypeptide or other polymer
  • modifications of non-natural amino acid components include, but are not limited to,
  • Such reactions are depicted in FIG. 2 wherein the amino acid functionality (A), translationally incorporated (or otherwise incorporated) into a polypeptide, reacts with reactant (B) to yield a modified polypeptide.
  • Such reactions may further occur with the amino acid functionality (A) on a polymer (including, by way of example, a polynucleotide, a polynucleoside, a polysaccharide, or combinations thereof), wherein reaction with reactant (B) yields a modified polymer.
  • a polymer including, by way of example, a polynucleotide, a polynucleoside, a polysaccharide, or combinations thereof
  • reaction with reactant (B) yields a modified polymer.
  • components refers to nonnatural amino acids, nonnatural amino acid polypeptides, polymers which contain nonnatural amino acids, nucleic acid sequences which contain selector codons, nonnatural amino acid polypeptides linked to polymers, nonnatural amino acid polypeptides linked to polymers which contain nonnatural amino acids, nonnatural amino acid polypeptides linked to nucleic acid sequences, nonnatural amino acid polypeptides linked to nucleic acid sequences; each of which may independently be a part of, or incorporated into, a polypeptide, a nonnatural amino acid polypeptide, nucleic acid sequence, or a polymer.
  • Non-natural amino acids with electrophile-containing sidechains including, but not limited to carbonyl groups such as aldehydes, esters, thioesters and ketones, can be incorporated into polypeptides.
  • the incorporation of such non-natural amino acids with such electrophilic sidechains into polypeptides makes possible site-specific derivatization of this sidechain via nucleophilic attack of the carbonyl group.
  • the attacking nucleophile is a hydroxylamine
  • an oxime-derivatized polypeptide will be generated.
  • the methods for derivatizing and/or further modifying may be conducted with a polypeptide that has been purified prior to the derivatization step or after the derivatization step. Further, the derivatization step can occur under mildly acidic to slightly basic conditions, including by way of example, between a pH of about 2 to about 10, or between a pH of about 2 to about 8, or between a pH of about 4 to about 8.
  • oxime groups may be further modified, such as, by way of example only, formation of masked oxime groups (which can be readily converted into oxime groups), protected oxime groups (which upon deprotection can be readily converted into oxime groups available for other chemical reactions), or new oxime groups via oxime exchange reactions.
  • Non-limiting examples of such modified polypeptide oxime linkages are shown below:
  • non-natural amino acids containing hydroxylamine groups into polypeptides allows for reaction with a variety of electrophilic groups including, but not limited to, carbonyl group such as ketones, esters, thioesters and aldehydes.
  • the nucleophilicity of the hydroxylamine group permits it to react efficiently and selectively with a variety of molecules that contain carbonyl functionality, or other functional groups with similar chemical reactivity, under mild conditions in aqueous solution to form the corresponding oxime linkage.
  • This site-specific derivatization and/or further modifying of such sidechains via nucleophilic attack of the carbonyl group may be conducted with a polypeptide that has been purified prior to the derivatization step or after the derivatization step.
  • the derivatization step can occur under mildly acidic to slightly basic conditions, including by way of example, between a pH of about 2 to about 10, a pH of about 2 to about 8, or between a pH of about 4 to about 8.
  • oxime groups may be further modified, such as, by way of example only, formation of masked oxime groups (which can be readily converted into oxime groups), protected oxime groups (which upon deprotection can be readily converted into oxime groups available for other chemical reactions), or new oxime groups via oxime exchange reactions.
  • Non-limiting examples of such modified polypeptide oxime linkages are shown below:
  • Non-natural amino acids containing an oxime group allow for reaction with a variety of reagents that contain certain reactive carbonyl groups (including but not limited to, aldehydes, esters, thioesters and ketones) to form new non-natural amino acids (which can be incorporated into a polypeptide) comprising a new oxime group.
  • Such an oxime exchange reaction allows for the further functionalization of non-natural amino acid polypeptides.
  • oxime groups may be further modified, such as, by way of example only, formation of masked oxime groups (which can be readily converted into oxime groups), protected oxime groups (which upon deprotection can be readily converted into oxime groups available for other chemical reactions), or new oxime groups via oxime exchange reactions.
  • Non-natural amino acids with electrophile-containing sidechains including, but not limited to dicarbonyl groups such as a diketone group, a ketoaldehyde group, a ketoacid group, a ketoester group, and a ketothioester group), a dicarbonyl-like group (which has reactivity similar to a dicarbonyl group and is structurally similar to a carbonyl group), a masked dicarbonyl group (which can be readily converted into a dicarbonyl group), or a protected dicarbonyl group (which has reactivity similar to a dicarbonyl group upon deprotection), can be incorporated into polypeptides.
  • dicarbonyl groups such as a diketone group, a ketoaldehyde group, a ketoacid group, a ketoester group, and a ketothioester group
  • a dicarbonyl-like group which has reactivity similar to a dicarbonyl group and is structurally similar to a
  • the incorporation of such unnatural amino acid with such electrophilic sidechains into polypeptides makes possible site-specific derivatization of this sidechain via nucleophilic attack of the carbonyl group.
  • the attacking nucleophile is a hydroxylamine
  • an oxime-derivatized polypeptide will be generated.
  • the methods for derivatizing and/or further modifying may be conducted with a polypeptide that has been purified prior to the derivatization step or after the derivatization step. Further, the derivatization step can occur under mildly acidic to slightly basic conditions, including by way of example, between a pH of about 2 to about 10, a pH of about 2 to about 8, or between a pH of about 4 to about 8.
  • oxime groups may be further modified, such as, by way of example only, formation of masked oxime groups (which can be readily converted into oxime groups), protected oxime groups (which upon deprotection can be readily converted into oxime groups available for other chemical reactions), or new oxime groups via oxime exchange reactions.
  • Non-limiting examples of such modified polypeptide oxime linkages are shown below:
  • non-natural amino acids containing hydroxylamine groups into polypeptides allows for reaction with a variety of electrophilic groups including, but not limited to, dicarbonyl group such as a diketone group, a ketoaldehyde group, a ketoacid group, a ketoester group, and a ketothioester group, a dicarbonyl-like group (which has reactivity similar to a dicarbonyl group and is structurally similar to a carbonyl group), a masked dicarbonyl group (which can be readily converted into a dicarbonyl group), or a protected dicarbonyl group (which has reactivity similar to a dicarbonyl group upon deprotection).
  • dicarbonyl group such as a diketone group, a ketoaldehyde group, a ketoacid group, a ketoester group, and a ketothioester group
  • a dicarbonyl-like group which has reactivity similar to a dicarbonyl group and is
  • the nucleophilicity of the hydroxylamine group permits it to react efficiently and selectively with a variety of molecules that contain such dicarbonyl functionality, or other functional groups with similar chemical reactivity, under mild conditions in aqueous solution to form the corresponding oxime linkage.
  • This site-specific derivatization and/or further modifying of such sidechains via nucleophilic attack of the dicarbonyl group may be conducted with a polypeptide that has been purified prior to the derivatization step or after the derivatization step.
  • the derivatization step can occur under mildly acidic to slightly basic conditions, including by way of example, between a pH of about 2 to about 10, a pH of about 2 to about 8, or between a pH of about 4 to about 8.
  • oxime groups may be further modified, such as, by way of example only, formation of masked oxime groups (which can be readily converted into oxime groups), protected oxime groups (which upon deprotection can be readily converted into oxime groups available for other chemical reactions), or new oxime groups via oxime exchange reactions.
  • Non-limiting examples of such modified polypeptide oxime linkages are shown below:
  • Non-natural amino acids containing an oxime group allow for reaction with a variety of reagents that contain certain reactive dicarbonyl groups, including, but not limited to, diketone groups, ketoaldehyde groups, ketoacid groups, ketoester groups, ketothioester groups, dicarbonyl-like groups (which has reactivity similar to a dicarbonyl group and is structurally similar to a carbonyl group), masked dicarbonyl groups (which can be readily converted into a dicarbonyl group), or protected dicarbonyl groups (which has reactivity similar to a dicarbonyl group upon deprotection) to form new non-natural amino acids (which can be incorporated into a polypeptide) comprising a new oxime group.
  • Such an oxime exchange reaction allows for the further functionalization of non-natural amino acid polypeptides.
  • oxime groups may be further modified, such as, by way of example only, formation of masked oxime groups (which can be readily converted into oxime groups), protected oxime groups (which upon deprotection can be readily converted into oxime groups available for other chemical reactions), or new oxime groups via oxime exchange reactions.
  • molecules can also be fused to the non-natural amino acid polypeptides described herein to modulate the half-life in serum.
  • molecules are linked or fused to the (modified) non-natural amino acid polypeptides described herein to enhance affinity for endogenous serum albumin in an animal.
  • a recombinant fusion of a polypeptide and an albumin binding sequence is made.
  • the (modified) non-natural amino acid polypeptides described herein are acylated with fatty acids.
  • the (modified) non-natural amino acid polypeptides described herein are fused directly with serum albumin (including but not limited to, human serum albumin).
  • serum albumin including but not limited to, human serum albumin.
  • Those of skill in the art will recognize that a wide variety of other molecules can also be linked to non-natural amino acid polypeptides, modified or unmodified, as described herein, to modulate binding to serum albumin or other serum components. Further discussion regarding the enhancement affinity for serum albumin is described in U.S. Patent Application Nos.
  • the methods and compositions described herein include polypeptides incorporating one or more non-natural amino acids bearing saccharide residues.
  • the saccharide residues may be either natural (including but not limited to, N-acetylglucosamine) or non-natural (including but not limited to, 3-fluorogalactose).
  • the saccharides may be linked to the non-natural amino acids either by an N- or O-linked glycosidic linkage (including but not limited to, N-acetylgalactose-L-serine) or a non-natural linkage (including but not limited to, an oxime or the corresponding C- or S-linked glycoside).
  • the saccharide (including but not limited to, glycosyl) moieties can be added to the non-natural amino acid polypeptides either in vivo or in vitro.
  • a polypeptide comprising a carbonyl-containing non-natural amino acid is modified with a saccharide derivatized with an aminooxy group to generate the corresponding glycosylated polypeptide linked via an oxime linkage.
  • the saccharide may be further elaborated by treatment with glycosyltransferases and other enzymes to generate an oligosaccharide bound to the non-natural amino acid polypeptide. See, e.g., H. Liu, et al. J. Am. Chem. Soc. 125: 1702-1703 (2003 ).
  • the non-natural amino acid portion of the polypeptide may first be modified with a multifunctional (e . g ., bi-, tri, tetra-) linker molecule that then subsequently is further modified. That is, at least one end of the multifunctional linker molecule reacts with at least one non-natural amino acid in a polypeptide and at least one other end of the multifunctional linker is available for further functionalization. If all ends of the multifunctional linker are identical, then (depending upon the stoichiometric conditions) homomultimers of the non-natural amino acid polypeptide may be formed.
  • a multifunctional linker molecule e . g ., bi-, tri, tetra-
  • the ends of the multifunctional linker have distinct chemical reactivities, then at least one end of the multifunctional linker group will be bound to the non-natural amino acid polypeptide and the other end can subsequently react with a different functionality, including by way of example only: a label; a dye; a polymer; a water-soluble polymer; a derivative of polyethylene glycol; a photocrosslinker; a cytotoxic compound; a drug; an affinity label; a photoaffinity label; a reactive compound; a resin; a second protein or polypeptide or polypeptide analog; an antibody or antibody fragment; a metal chelator; a cofactor; a fatty acid; a carbohydrate; a polynucleotide; a DNA; a RNA; an antisense polynucleotide; a saccharide; a water-soluble dendrimer; a cyclodextrin; an inhibitory ribonucleic acid; a biomaterial; a nanoparticle;
  • a naturally-occurring or non-natural amino acid polypeptide may be difficult to isolate from a sample for a number of reasons, including but not limited to the solubility or binding characteristics of the polypeptide.
  • a polypeptide in the preparation of a polypeptide for therapeutic use, such a polypeptide may be isolated from a recombinant system that has been engineered to overproduce the polypeptide.
  • solubility or binding characteristics of the polypeptide achieving a desired level of purity often proves difficult.
  • a naturally-occurring or non-natural amino acid polypeptide may be difficult to detect in a sample (including an in vivo sample and an in vitro sample) for a number of reasons, including but not limited to the lack of a reagent or label that can readily bind to the polypeptide.
  • the methods, compositions, techniques and strategies further described in U.S. Patent Application Nos. 60/638,418 , 60/638,527 , 60/639,195 , 60/696,210 , 60/696,302 , and 60/696,068 ; PCT Publication WO 05/074650 entitled "Modified Four Helical Bundle Polypeptides and Their Uses," in their entirety provide a solution to this situation.
  • a naturally-occurring or non-natural amino acid polypeptide will be able to provide a certain therapeutic benefit to a patient with a particular disorder, disease or condition. Such a therapeutic benefit will depend upon a number of factors, including by way of example only: the safety profile of the polypeptide, and the pharmacokinetics, pharmacologics and/or pharmacodynamics of the polypeptide (e . g ., water solubility, bioavailability, serum half-life, therapeutic half-life, immunogenicity, biological activity, or circulation time).
  • the (modified) non-natural amino acid polypeptides described herein, including homo- and hetero-multimers thereof find multiple uses, including but not limited to: therapeutic, diagnostic, assay-based, industrial, cosmetic, plant biology, environmental, energy-production, and/or military uses.
  • therapeutic, diagnostic, assay-based industrial, cosmetic, plant biology, environmental, energy-production, and/or military uses.
  • the following therapeutic uses of (modified) non-natural amino acid polypeptides are provided.
  • the (modified) non-natural amino acid polypeptides described herein are useful for treating a wide range of disorders.
  • Administration of the (modified) non-natural amino acid polypeptide products described herein results in any of the activities demonstrated by commercially available polypeptide preparations in humans.
  • Average quantities of the (modified) non-natural amino acid polypeptide product may vary and in particular should be based upon the recommendations and prescription of a qualified physician.
  • the exact amount of the (modified) non-natural amino acid polypeptide is a matter of preference subject to such factors as the exact type of condition being treated, the condition of the patient being treated, as well as the other ingredients in the composition.
  • the amount to be given may be readily determined by one skilled in the art based upon therapy with the (modified) non-natural amino acid polypeptide.
  • non-natural amino acid polypeptides, modified or unmodified, as described herein are optionally employed for therapeutic uses, including but not limited to, in combination with a suitable pharmaceutical carrier.
  • Such compositions for example, comprise a therapeutically effective amount of the non-natural amino acid polypeptides, modified or unmodified, as described herein, and a pharmaceutically acceptable carrier or excipient.
  • a carrier or excipient includes, but is not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and/or combinations thereof.
  • the formulation is made to suit the mode of administration. In general, methods of administering proteins are well known in the art and can be applied to administration of the non-natural amino acid polypeptides, modified or unmodified, as described herein.
  • compositions comprising one or more of the non-natural amino acid polypeptides, modified or unmodified, as described herein are optionally tested in one or more appropriate in vitro and/or in vivo animal models of disease, to confirm efficacy, tissue metabolism, and to estimate dosages, according to methods known to those of ordinary skill in the art.
  • dosages can be initially determined by activity, stability or other suitable measures of non-natural to natural amino acid homologues (including but not limited to, comparison of a polypeptide (modified) to include one or more non-natural amino acids to a natural amino acid polypeptide), i. e., in a relevant assay.
  • Administration is by any of the routes normally used for introducing a molecule into ultimate contact with blood or tissue cells.
  • the non-natural amino acid polypeptides, modified or unmodified, as described herein, are administered in any suitable manner, optionally with one or more pharmaceutically acceptable carriers.
  • Suitable methods of administering the non-natural amino acid polypeptides, modified or unmodified, as described herein, to a patient are available, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective action or reaction than another route.
  • compositions are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions described herein.
  • Non-natural amino acid polypeptides may be administered by any conventional route suitable for proteins or peptides, including, but not limited to parenterally, e.g. injections including, but not limited to, subcutaneously or intravenously or any other form of injections or infusions.
  • Polypeptide compositions (including the various polypeptides described herein) can be administered by a number of routes including, but not limited to oral, intravenous, intraperitoneal, intramuscular, transdermal, subcutaneous, topical, sublingual, or rectal means.
  • Compositions comprising non-natural amino acid polypeptides, modified or unmodified, as described herein can also be administered via liposomes. Such administration routes and appropriate formulations are generally known to those of skill in the art.
  • the non-natural amino acid polypeptide may be used alone or in combination with other suitable components such as a pharmaceutical carrier.
  • non-natural amino acid polypeptides modified or unmodified, as described herein, alone or in combination with other suitable components, can also be made into aerosol formulations ( i . e ., they can be "nebulized") to be administered via inhalation. Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • the formulations of packaged nucleic acid can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials.
  • Parenteral administration and intravenous administration are preferred methods of administration.
  • the routes of administration already in use for natural amino acid homologue therapeutics including but not limited to, those typically used for EPO, IFN, GM-CSF, IFNs, interleukins, antibodies, and/or any other pharmaceutically delivered protein
  • formulations in current use provide preferred routes of administration and formulation for the non-natural amino acid polypeptides, modified or unmodified, as described herein.
  • the dose administered to a patient is sufficient to have a beneficial therapeutic response in the patient over time.
  • the dose is determined by the efficacy of the particular formulation, and the activity, stability or serum half-life of the non-natural amino acid polypeptides, modified or unmodified, employed and the condition of the patient, as well as the body weight or surface area of the patient to be treated.
  • the size of the dose is also determined by the existence, nature, and extent of any adverse side-effects that accompany the administration of a particular formulation, or the like in a particular patient.
  • the physician evaluates circulating plasma levels, formulation toxicities, progression of the disease, and/or where relevant, the production of anti-non-natural amino acid polypeptide antibodies.
  • the dose administered, for example, to a 70 kilogram patient is typically in the range equivalent to dosages of currently-used therapeutic proteins, adjusted for the altered activity or serum half-life of the relevant composition.
  • the pharmaceutical formulations described herein can supplement treatment conditions by any known conventional therapy, including antibody administration, vaccine administration, administration of cytotoxic agents, natural amino acid polypeptides, nucleic acids, nucleotide analogues, biologic response modifiers, and the like.
  • the pharmaceutical formulations described herein are administered at a rate determined by the LD-50 or ED-50 of the relevant formulation, and/or observation of any side-effects of the non-natural amino acid polypeptides, modified or unmodified, at various concentrations, including but not limited to, as applied to the mass and overall health of the patient. Administration can be accomplished via single or divided doses.
  • a patient undergoing infusion of a formulation develops fevers, chills, or muscle aches, he/she receives the appropriate dose of aspirin, ibuprofen, acetaminophen or other pain/fever controlling drug.
  • Patients who experience reactions to the infusion such as fever, muscle aches, and chills are premedicated 30 minutes prior to the future infusions with either aspirin, acetaminophen, or, including but not limited to, diphenhydramine.
  • Meperidine is used for more severe chills and muscle aches that do not quickly respond to antipyretics and antihistamines. Cell infusion is slowed or discontinued depending upon the severity of the reaction.
  • Non-natural amino acid polypeptides, modified or unmodified, as described herein can be administered directly to a mammalian subject. Administration is by any of the routes normally used for introducing a polypeptide to a subject.
  • the non-natural amino acid polypeptides, modified or unmodified, as described herein include those suitable for oral, rectal, topical, inhalation (including but not limited to, via an aerosol), buccal (including but not limited to, sub-lingual), vaginal, parenteral (including but not limited to, subcutaneous, intramuscular, intradermal, intraarticular, intrapleural, intraperitoneal, inracerebral, intraarterial, or intravenous), topical (i.e., both skin and mucosal surfaces, including airway surfaces) and transdermal administration, although the most suitable route in any given case will depend on the nature and severity of the condition being treated.
  • Administration can be either local or systemic.
  • the formulations can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials.
  • the non-natural amino acid polypeptides, modified or unmodified, as described herein can be prepared in a mixture in a unit dosage injectable form (including but not limited to, solution, suspension, or emulsion) with a pharmaceutically acceptable carrier.
  • the non-natural amino acid polypeptides, modified or unmodified, as described herein can also be administered by continuous infusion (using, including but not limited to, minipumps such as osmotic pumps), single bolus or slow-release depot formulations.
  • Formulations suitable for administration include aqueous and non-aqueous solutions, isotonic sterile solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. Solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • Freeze-drying is a commonly employed technique for presenting proteins which serves to remove water from the protein preparation of interest.
  • Freeze-drying or lyophilization, is a process by which the material to be dried is first frozen and then the ice or frozen solvent is removed by sublimation in a vacuum environment.
  • An excipient may be included in pre-lyophilized formulations to enhance stability during the freeze-drying process and/or to improve stability of the lyophilized product upon storage. Pikal, M. Biopharm. 3(9)26-30 (1990 ) and Arakawa et al. Pharm. Res. 8(3):285-291 (1991 ).
  • Spray drying of pharmaceuticals is also known to those of ordinary skill in the art. For example, see Broadhead, J. et al., "The Spray Drying of Pharmaceuticals," in Drug Dev. Ind. Pharm, 18 (11 & 12), 1169-1206 (1992 ).
  • a variety of biological materials have been spray dried and these include: enzymes, sera, plasma, micro-organisms and yeasts.
  • Spray drying is a useful technique because it can convert a liquid pharmaceutical preparation into a fine, dustless or agglomerated powder in a one-step process.
  • the basic technique comprises the following four steps: a) atomization of the feed solution into a spray; b) spray-air contact; c) drying of the spray; and d) separation of the dried product from the drying air.
  • U.S. Patent Nos. 6,235,710 and 6,001,800 describe the preparation of recombinant erythropoietin by spray drying.
  • compositions described herein may comprise a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions (including optional pharmaceutically acceptable carriers, excipients, or stabilizers) for the non-natural amino acid polypeptides, modified or unmodified, described herein, ( see , e.g ., Remington's Pharmaceutical Sciences, 17th ed. 1985 )).
  • Suitable carriers include buffers containing succinate, phosphate, borate, HEPES, citrate, imidazole, acetate, bicarbonate, and other organic acids; antioxidants including but not limited to, ascorbic acid; low molecular weight polypeptides including but not limited to those less than about 10 residues; proteins, including but not limited to, serum albumin, gelatin, or immunoglobulins; hydrophilic polymers including but not limited to, polyvinylpyrrolidone; amino acids including but not limited to, glycine, glutamine, asparagine, arginine, histidine or histidine derivatives, methionine, glutamate, or lysine; monosaccharides, disaccharides, and other carbohydrates, including but not limited to, trehalose, sucrose, glucose, mannose, or dextrins; chelating agents including but not limited to, EDTA; divalent metal ions including but not limited to, zinc, cobalt, or copper; sugar alcohols including but not limited
  • Suitable surfactants include for example but are not limited to polyethers based upon poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide), i.e., (PEO-PPO-PEO), or poly(propylene oxide)-poly(ethylene oxide)-poly(propylene oxide), i.e., (PPO-PEO-PPO), or a combination thereof.
  • PEO-PPO-PEO and PPO-PEO-PPO are commercially available under the trade names Pluronics TM , R-Pluronics TM , Tetronics TM and R-Tetronics TM (BASF Wyandotte Corp., Wyandotte, Mich.) and are further described in U.S. Pat. No.
  • ethylene/polypropylene block polymers may be suitable surfactants.
  • a surfactant or a combination of surfactants may be used to stabilize a (modified) non-natural amino acid polypeptide against one or more stresses including but not limited to stress that results from agitation. Some of the above may be referred to as “bulking agents.” Some may also be referred to as “tonicity modifiers.”
  • the non-natural amino acid polypeptides, modified or unmodified, as described herein, including those linked to water soluble polymers such as PEG can also be administered by or as part of sustained-release systems.
  • Sustained-release compositions include, including but not limited to, semi-permeable polymer matrices in the form of shaped articles, including but not limited to, films, or microcapsules.
  • Sustained-release matrices include from biocompatible materials such as poly(2-hydroxyethyl methacrylate) ( Langer et al., J. Biomed. Mater. Res., 15: 267-277 (1981 ); Langer, Chem.
  • polyglycolide polymer of glycolic acid
  • polylactide co-glycolide copolymers of lactic acid and glycolic acid
  • polyanhydrides copolymers of L-glutamic acid and gamma-ethyl-L-glutamate ( Sidman et al., Biopolymers, 22, 547-556 (1983 ), poly(ortho)esters, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, polyamino acids, amino acids such as phenylalanine, tyrosine, isoleucine, polynucleotides, polyvinyl propylene, polyvinylpyrrolidone and silicone.
  • Sustained-release compositions also include a liposomally entrapped compound.
  • Liposomes containing the compound are prepared by methods known per se: DE 3,218,121 ; Eppstein et al., Proc. Natl. Acad. Sci. U.S.A., 82: 3688-3692 (1985 ); Hwang et al., Proc. Natl. Acad. Sci. U.S.A., 77: 4030-4034 (1980 ); EP 52,322 ; EP 36,676 ; U.S. Patent No. 4,619,794 ; EP 143,949 ; U.S. Patent No. 5,021,234 ; Japanese Pat. Appln. 83-118008 ; U.S. Pat. Nos. 4,485,045 and 4,544,545 ; and EP 102,324 .
  • Liposomally entrapped polypeptides can be prepared by methods described in, e.g., DE 3,218,121 ; Eppstein et al., Proc. Natl. Acad. Sci. U.S.A., 82: 3688-3692 (1985 ); Hwang et al., Proc. Natl. Acad. Sci. U.S.A., 77: 4030-4034 (1980 ); EP 52,322 ; EP 36,676 ; U.S. Patent No. 4,619,794 ;; EP 143,949 ; U.S. Patent No. 5,021,234 ; Japanese Pat. Appln. 83-118008 ; U.S. Patent Nos.
  • composition and size of liposomes are well known or able to be readily determined empirically by one of ordinary skill in the art. Some examples of liposomes as described in, e.g., Park JW, et al., Proc. Natl. Acad. Sci.
  • the dose administered to a patient in the context of the compositions, formulations and methods described herein, should be sufficient to cause a beneficial response in the subject over time.
  • the total pharmaceutically effective amount of the non-natural amino acid polypeptides, modified or unmodified, as described herein, administered parenterally per dose is in the range of about 0.01 ⁇ g/kg/day to about 100 ⁇ g/kg, or about 0.05 mg/kg to about 1 mg/kg, of patient body weight, although this is subject to therapeutic discretion.
  • the frequency of dosing is also subject to therapeutic discretion, and may be more frequent or less frequent than the commercially available products approved for use in humans.
  • a polymer:polypeptide conjugate including by way of example only, a PEGylated polypeptide, as described herein, can be administered by any of the routes of administration described above.
  • the isolation of peptides, (modified) non-natural amino acid polypeptides, binding partners or receptors to polypeptides can occur by chromatography. Chromatography is based on the differential absorption and elution of polypeptides.
  • the sample is dissolved in a mobile phase, which may be a gas, a liquid or a supercritical fluid. This mobile phase is then forced through an immiscible stationary phase, which is fixed in a column or on a solid surface.
  • stationary phases include liquids adsorbed on a solid, organic species bonded to a solid surface, solid, ion exchange resin and liquid in interstices of a polymeric solid.
  • the ability of a polypeptide to be purified by different chromatographic or other isolation/purification methods may be modulated by the addition or substitution of one or more non-natural amino acids with a non-natural amino acid optionally in combination with one or more natural amino acid substitutions.
  • the properties of a polypeptide may be modified by altereing the amino acid composition enabling an increase or decrease in its interaction with known matrices. Changes to the amino acid composition include, but are not limited to, hydrophobic amino acid content, hydrophilic amino acid content, and change in charge, pI, or other characteristics of the polypeptide. Such modifications may be useful in isolating membrane proteins which are difficult to isolate since they are hydrophobic in nature and keep in their native conformation.
  • the isolation of polypeptides can occur by gas chromatography (GC).
  • GC gas chromatography
  • the sample is vaporized and injected onto the head of a chromatographic column.
  • mobile gas phases include but are not limited to helium, argon, nitrogen, carbon dioxide, and hydrogen.
  • the sample is isolated by gas-solid chromatography, where the stationary phase is a solid.
  • solid stationary phase are molecular sieves and porous polymers.
  • the polypeptide is isolated by gas-liquid chromatography, where the stationary phase is a liquid immobilized on the surface of an inert solid.
  • liquid stationary phases examples include Polydimethyl siloxane, Poly (phenylmethyldimethyl) siloxane (10% phenyl), Poly(phenylmethyl) siloxane (50% phenyl), Poly(trifluoropropyldimethyl) siloxane, Polyethylene glycol and Poly(dicyanoallyldimethyl) siloxane.
  • GC columns are either packed and open tubular or capillary.
  • GC-chromatographic columns vary in length from less than 2 m to 50 m or more. Examples of material for their construction include stainless steel, metal, glass, fused silica and Teflon.
  • Typically GC columns have an in inner diameter of roughly of 2 to 4 mm.
  • Micro-GC has an inner diameter of roughly 1 mm.
  • Capillary GC utilizes a capillary with an inner diameter of roughly 100 to 750 um.
  • Nano-GC is available with an inner diameter of 50 um -1 mm
  • LC liquid chromatography
  • LC columns are ordinarily constructed from smooth-bore stainless steel tubing, although heavy glass tubing in occasionally encountered.
  • Conventional LC columns have an inner diameter of roughly 4.6 mm and a flow rate of roughly 1 ml/min.
  • Micro-LC has an inner diameter of roughly 1.0 mm and a flow rate of roughly 40 ⁇ l/min.
  • Capillary LC utilizes a capillary with an inner diameter of roughly 300 ⁇ m and a flow rate of approximately 5 ⁇ l/min.
  • Nano-LC is available with an inner diameter of 50 ⁇ m -1 mm and flow rates of 200 nl/min. Nano-LC can vary in length, e.g., 5, 15, or 25 cm. Nano-LC stationary phase may also be a monolithic material, such as a polymeric monolith or a sol-gel monolith.
  • Two basic types of packing material have been used in liquid chromatography, non-porous and porous particles.
  • the beads or particles are generally characterized by particle and pore size. Particle sizes generally range between 3 and 50 microns. Larger particles will generate less system pressure and smaller particles will generate more pressure. The smaller particles generally give higher separation efficiencies.
  • the particle pore size is measured in angstroms and generally range between 100-1000 A. These can be covered with a porous layer of silica, alumina, ion exchange resin, organic surface layer, polymers, ligands, carbohydrates or a specific cofactor.
  • the polypeptides can be isolated using HPLC technology.
  • the polypeptide can be isolated using column chromatography. In column chromatography, the solid medium is packed onto a chromatography column, and the initial mixture containing the polypeptide is run through the column to allow binding. A wash buffer is then run through the column, and the elution buffer is subsequently applied to the column for sample collection. These steps may be performed at ambient pressure.
  • binding of the polypeptides to a solid phase may be achieved using a Batch treatment, by adding the initial mixture to the solid phase in a vessel, mixing the two together, separating the solid phase (i.e.
  • the isolation of peptides occur in a microfluidic device. In another instance, the isolation of peptides occur in a nanofluidic device.
  • the isolation of polypeptides occurs by partition chromatography. In one instance the isolation of the polypeptides occurs by liquid-liquid partition chromatography. With liquid-liquid partition chromatography, a liquid stationary phase is retained on the surface of the packing by physical adsorption. In another instance, the isolation of the polypeptides can occur by bonded-phase partition chromatography. With bonded-phase partition chromatography, the stationary phase is bonded chemically to the support surfaces.
  • normal-phase chromatography is used to isolate the polypeptides.
  • a polar stationary phase is used together with a non-polar solvent.
  • the stationary phase for normal phase chromatography include but are not limited to, water, alcohols and triethylene glycol.
  • non-polar solvents for normal phase chromatography include but are not limited to, ethyl, ether, chloroform, tetrahydrofuran, flouroalkanes, cyclohexane, 1-chlorobutane, carbon tetrachloride, toluene, diethyl ether, hexane and i-propylether.
  • the partition chromatography uses reversed-phase packings; this is referred as reversed-phase chromatography.
  • reversed-phase chromatography a non-polar stationary phase is used together with a polar mobile phase.
  • stationary phases for reversed-phased chromatography include but are not limited to, hydrocarbons, ether, esters, ketones, aldehydes, amides, and amines.
  • mobile stationary phases for reversed-phased chromatography include water, methanol, ethanol, ethyl acetate, dioxane, nitromethane, ethylene glycol, tetrahydrofuran and acetonitrile.
  • the type of reversed chromatography that can be use to isolate polypeptides is ion-pair chromatography.
  • the mobile phase in ion-pair chromatography consists of an aqueous buffer containing an organic solvent such as methanol or acetonitrile and an ionic compound containing a counter ion of opposite charge to the polypeptides.
  • the counter ion binds to the polypeptide to form an ion pair, which is a neutral species that is retained by a reversed-phase packing. Elution of the ion pairs is then accomplished with an aqueous solution of methanol or another water soluble organic solvent like the one described above.
  • Examples of counter-ions are ClO 4 - , C 12 H 25 SO 3 - , (C 4 H 9 ) 4 N + , (C 16 H 33 )(CH 3 ) 3 N + , (C 4 H 9 ) 4 N + , Bis-(2-ethylhexyl)phosphate, and (C 4 H 9 )4N + .
  • the polypeptides can be isolated using partition chromatography with a chiral stationary phase.
  • chiral stationary phases include but are not limited to, protein based stationary phases, small molecular weight chiral, polymers of cellulose and amylose, macrocyclic glycopeptides and cyclodextrin based materials.
  • the isolation of polypeptides can occur by adsorption chromatography.
  • Adsorption is a process whereby material (contained in the mobile phase) interacts by physical forces (dispersive, polar or ionic) with a stationary phase, thereby, causing a layer (or layers) of the material to adhere to that stationary phase.
  • the stationary phase in most cases will be a solid (e.g. silica gel, alumina, charcoal, etc.) or sometimes a liquid (e.g. surfactants on water surfaces).
  • the surface layer(s) may be single, double or multiple.
  • the isolation of polypeptides can occur by ion-exchange chromatography.
  • ion-exchange chromatography the isolation of polypeptides is based upon ion-exchange resin.
  • the ion exchange resin can be an anion exchange resin or a cation exchange resin.
  • the ion-exchange resin can be made by natural ion exchangers, such as clays and zeolites, or from synthetic ion exchangers. Examples of common active sites for cation exchange resins are the sulfonic acid group -SO 3 - H + , the carboxylic acid group -COO - H + and phosphoric acid -PO 32 + H 2 .
  • the mobile phase in ion-exchange chromatography is generally an aqueous solution that may contain moderate amounts of methanol or other water miscible organic solvents; these mobile phases also contain ionic species in the form of a buffer.
  • the ion exchange column is eluted with a gradient of salt concentrations.
  • pumps add increasing amounts of salt to the buffer as it goes onto the column so that there is a continuous steady increase in the ionic concentration going through the column.
  • the proteins then "elute" or come off the column stationary phase when the ionic strength of the buffer neutralizes their charge. The least charged molecules come off first, and the most highly charged come off last.
  • the column is thoroughly rinsed with buffers of increasing ionic strength until the desired protein elutes; this exact same sequence is repeated each time with the same amounts of buffer to give reproducible yields and purification of the protein.
  • the sample will be subject removal of high salt concentrations after isolation the polypeptide of interest by ion exchange chromatography.
  • the removal of high salt concentration will be performed by dialysis.
  • Dialysis makes use of semi-permeable membranes. The main feature of the dialysis membrane is that it is porous. However, the pore size is such that while small salt ions can freely pass through the membrane, larger protein molecules cannot (i.e. they are retained). Thus, dialysis membranes are characterized by the molecular mass of the smallest typical globular protein which it will retain. Removal of high salt concentration can be achieved in a single or multiple dialysis steps. In another embodiment, the removal of high salt concentration is performed by electrodialysis.
  • Electrodialysis is an electromembrane process in which ions are transported through ion permeable membranes from one solution to another under the influence of a potential gradient. Since the membranes used in electrodialysis have the ability to selectively transport ions having positive or negative charge and reject ions of the opposite charge, electrodialysis is useful for concentration, removal, or separation of electrolytes.
  • the removal of high concentration of salt is achieved by using desalting columns in gravity-flow gel filtration.
  • Gravity-flow gel filtration involves the chromatographic separation of molecules of different dimensions based on their relative abilities to penetrate into a suitable stationary phase.
  • Desalting columns are packed with small, porous cellulose beads. These columns have a wet bead with specific diameters. The diameter of the beads used will depend on the molecular weight of the peptide of interest. Different levels of separation can be achieved based on the pore size of the medium packed into the column.
  • the medium can be chosen to totally exclude proteins or large molecules, while still including small solutes. Large molecules are excluded from the internal pores of the gel and emerge first from the column. The smaller molecules are able to penetrate the pores, and then progress through the column at a slower rate. These smaller molecules are subsequently flushed through the column with additional buffer volume.
  • the isolation of polypeptides can occur by size-exclusion chromatography, also known as gel permeation, or gel filtration chromatography. Molecules that are larger than the average pore size of the packing are excluded and thus suffer no retention.
  • packings for size exclusion chromatography include silica, cellulose beads and polymer particles. Conventionally, porous glasses and silica particles have an average pore size ranging from 40 ⁇ to 2500 ⁇ . In some instance, the molecular weight exclusion limit of a polymer packing with an average pore size of 102 ⁇ is 700. In another instance the molecular weight exclusion limit of a polymer packing with an average pore size of 103 ⁇ is (0.1 to 20) x 10 4 .
  • the molecular weight exclusion limit of a polymer packing with an average pore size of 104 ⁇ is (1 to 20) x 10 4 .
  • the molecular weight exclusion limit of a polymer packing with an average pore size of 10 5 ⁇ is (1 to 20) x 10 5 .
  • the molecular weight exclusion limit of a polymer packing with an average pore size of 10 6 ⁇ is (5 to 10) ⁇ 10 6 .
  • the molecular weight exclusion limit of silica packing with an average pore size of 125 ⁇ is (0.2 to 5) x 10 4 .
  • the molecular weight exclusion limit of silica packing with an average pore size of 300 ⁇ is (0.03 to 1) x 10 5 .
  • the molecular weight exclusion limit of silica packing with an average pore size of 500 ⁇ is (0.05 to 5) x 10 5 .
  • the molecular weight exclusion limit of silica packing with an average pore size of 1000 ⁇ is (5 to 20) x 10 5 .
  • the isolation of polypeptides can occur by thin-layer chromatography.
  • Thin-layer chromatographic methods include paper chromatography, thin-layer chromatography and electrochromatography.
  • Each makes use of a flat, thin layer of material that is either self supporting or that is coated on a glass, plastic, or metal surface.
  • the mobile phase moves through the stationary phase by capillary action, sometimes assisted by gravity or electrical potential.
  • planar separation is performed on flat glass or plastic plates that are coated with a thin and adherent layer of finely divided particles; this layer constitutes the stationary phase.
  • the stationary phase and mobile phase are similar to those discussed in adsorption, normal- and reversed-phase partition, ion-exchange, and size exclusion chromatography.
  • the polypeptides are located in the plate by spraying a solution that will react with organic compounds to yield dark products. Examples of this type of solution include ninhydrin, iodine solutions and sulfuric acid solution.
  • the polypeptides are located by incorporating a fluorescent material to the stationary phase. The plate is examined under ultraviolet light. The sample components quench the fluorescent material so that all of the plate fluoresces except where the non-fluorescing sample components are located.
  • the isolation of polypeptides can occur by affinity chromatography.
  • Affinity chromatography relies on the ability to design a stationary phase that reversibly binds to a known subset of molecules.
  • Affinity purification generally involves the following steps: 1) incubate crude sample with the immobilized ligand support material to allow the target molecule in the sample to bind to the immobilized ligand, 2) wash away nonbound sample components from solid support and 3) elute (dissociate and recover) the target molecule from the immobilized ligand by altering the buffer conditions so that the binding interaction no longer occurs.
  • elution buffers used in affinity chromatography include but are not limited to 100 mM glycine•HCl, 100 mM citric acid, 50-100 mM triethylamine or triethanolamine, 150 mM ammonium hydroxide, 3.5-4.0 M magnesium chloride in 10 mM Tris, 5 M lithium chloride in 10 mM phosphate buffer, 2.5 M sodium iodide, 0.2-3.0 sodium thiocyanate, 2-6 M guanidine•HCl, 2-8 M urea, 1% deoxycholate, 1 % SDS , 10% dioxane, 50% ethylene glycol, 0.1 M Glycine-NaOH, 0.1 M Glycine-NaOH with 50% ethyleneglycol, 3.0 M Potassium chloride, 0.1 M Tris-acetate with 2.0 M NaCl, 5.0 M Potassium iodide, 1% SDS, 1% Sodium deoxycholate, 2.0 M Urea, 6.0 M Urea,
  • the stationary phase includes a ligand including but not limited to, a specific carbohydrate or a cofactor.
  • the polypeptides can then be eluted with a high concentration of the carbohydrate or a specific cofactor. Mimics for binding sites can sometimes be used as affinity stationary phases.
  • the specific sugars, inhibitor or cofactors used in the stationary phase will vary according to the properties of the polypeptide and include any ligand, carbohydrate or cofactor known in the art.
  • the immobilized stationary phase includes a dye.
  • dyes commonly used for dye-ligand chromatography include Reactive Blue 2 (Cibacron ⁇ Blue 3GA), Reactive Red 120 (Procion ⁇ Red HE3B), Reactive Blue 4 (Reactive Blue MRB ) TC , Reactive Green 5 (Reactive Green H4G) TC , Reactive Green 19 (Reactive Green HE4BD) TC , Green 19A (Reactive Green HE4BD) TC , Reactive Yellow 86 (Reactive Yellow M8G) TC and Reactive Brown 10 (Reactive Brown M4R ) TC .
  • the stationary phase includes a metal chelate resin.
  • metal chelate chromatography metal ions such as Zn 2+ , Cu 2+ and Ni 2+ are immobilized to a chromatography stationary phase by chelate bonding take part in a reversible interaction with electron donor groups situated in the surface of polypeptides.
  • the polypeptide is bonded to the stationary phase and can be subsequently eluted by means of a buffer with lower pH value at which the electron group is protonized.
  • chelate resins include 8-hydroxyquinoline, salicylic acid, diethylenetriamine, diethylenetriaminetetraacetic acid, ethylenediaminetetraacetic acid (EDTA), iminodiacetic acid and nitrilo-triacetic acid.
  • the isolation of polypeptides can occur by immunoaffinity chromatography.
  • the principle of immunoaffinity or immunoadsorption chromatography is based on the highly specific interaction of an antigen with its antibody.
  • Immunoaffinity chromatography utilizes an antibody or antibody fragment as a ligand immobilized onto the stationary phase in a manner that retains its binding capacity. Elution of the retained polypeptide is achieved by alterations to the mobile-phase conditions that weaken the antibody-antigen interaction. Elution conditions are intended to break the ionic, hydrophobic and hydrogen bonds that hold the antigen and antibody together. Successful eluting conditions will be dependent upon the specific antigen-antibody interaction that is occurring.
  • Antibodies may be generated that recognize the non-natural amino acid present in the polypeptide. Such antibodies may be used in affinity chromatography to purify the non-natural amino acid polypeptides from a complex mixture or enable conjugation of the polypeptide with other molecules on a support such as a resin, in immunoassays to detect the presence of non-natural amino acid polypeptides, and other assays that use antibodies. Antibodies may be generated that recognize one or more non-natural amino acids present at the N or C terminus of a polypeptide or other portions of the polypeptide.
  • Non-natural amino acid polypeptides may be antibodies, antibody fragments, or antigen-binding polypeptides or fragments thereof, and used to isolate antigens by affinity chromatography.
  • polypeptides can occur by hydrophobic-interaction chromatography.
  • Polyeptides may contain hydrophilic and hydrophobic natural amino acids and hydrophilic and hydrophobic non-natural amino acids. Polypeptides are separated according to their relative hydrophobicity by their ability to reversibly bind to hydrophobic compounds. The polypeptides are eluted from the column with decreasing concentrations of salt in buffer. Examples of hydrophobic compounds include but are not limited to, hydrophobic fatty acid chains, compounds with n-butyl functional groups, compounds with n-octyl functional groups and compounds with phenyl functional groups.
  • the isolation of polypeptides can occur by supercritical chromatography (SFC).
  • SFC supercritical chromatography
  • the sample is carried through a separating column by a supercritical fluid where the mixture is divided into unique bands based on the amount of interaction between the individual analytes and the stationary phase in the column.
  • Conventional SFC columns are either packed and open tubular or capillary. Open-tubular columns vary in length from 10 m to 20 m or more. Typically open-tubular columns have an inner diameter of roughly of 0.05 to 4 mm. Pack columns vary in diameter from 0.5 mm or less to 4.6 mm, with particle diameter ranging from 3 to 10 um. Packed columns contain small deactivated particles to which the stationary phases adhere.
  • the columns are conventionally stainless steel.
  • Capillary columns are open tubular columns of narrow internal diameter made of fused silica, with the stationary phase bonded to the wall of the column.
  • the coatings are similar to those used in partition chromatography.
  • supercritical fluids used in SFC include but are not limited to, carbon dioxide, ethane, pentane, nitrous oxide, dichlorodifluoromethane, diethyl ether, ammonia, and tetrahydrofuran.
  • polar organic modifiers such as methanol are introduced in small concentrations (1-5%).
  • the isolation of peptides, (modified) non-natural amino acid polypeptides, binding partners or receptors to polypeptides can occur by precipitation.
  • the solubility of polypeptides is a function of the ionic strength and pH of the solution.
  • Polypeptides have isoelectric points at which the charges of their amino acid side groups balance each other. If the ionic strength of a solution is either very high or very low, proteins will tend to precipitate at their isoelectric point. In one embodiment, the ionic strength of the solution will be increased by adding salt. Examples of salts used in precipitation methods include but are not limited to ammonium sulfate and sodium sulfate.
  • any salt known in the art for protein precipitation can be used in any of the embodiments of the inventions.
  • polypeptides will be forced out of solution with polymers.
  • a polymer commonly used to precipitate polypeptides is polyethylene glycol.
  • Any polymer known in the art for protein precipitation can be used in any of the embodiments of the inventions.
  • the precipitated polypeptides are removed by centrifugation or filtration.
  • the sample after precipitation of the peptide of interest by the addition salts to the solution the sample will be subject removal of high salt concentrations. Desalting methods are discussed in the ion-exchange chromatography section.
  • IP immunoprecipitation
  • Classical immunoprecipitation involves the following steps: 1)incubate specific antibody with a sample containing antigen, 2) capture antibody-antigen complex with immobilized Protein A or G agarose gel (Protein A or G binds the antibody, which is bound to its antigen), 3)Wash the gel with buffer to remove non-bound sample components, 4)Elute the antigen (and antibody).
  • modifications to the classical IP method can be made so that the antibody is permanently immobilized and will not elute with the antigen.
  • the antibody is first bound to the Protein A or G gel and then the antibody is covalently cross-linked to the Protein A or G.
  • the antibody is directly coupled to an activated affinity support.
  • Non-natural amino acid polypeptides may be antigen-binding polypeptides and used in immunoprecipitation.
  • the support material is a porous gel such as cross-linked beaded agarose or co-polymer of cross-linked bis-acrylamide and azlactone.
  • polypeptides can be isolated by magnetic affinity separation. Samples containing the molecule of interest are incubated with magnetic beads that are derivatized with an antibody or other binding partner. A magnetic field is used to pull the magnetic beads out of solution and onto a surface. The buffer can be carefully removed, containing any nonbound molecules. Protocols using magnetic beads for isolation of molecules of interest are well known in the art.
  • Magnetic beads can be derivatized to contain active groups, including but not limited to, carboxylic acids or primary amines, or specific affinity molecules such as streptavidin or goat anti-mouse, anti-rabbit or anti-rat IgG or Protein A or G.
  • active groups including but not limited to, carboxylic acids or primary amines, or specific affinity molecules such as streptavidin or goat anti-mouse, anti-rabbit or anti-rat IgG or Protein A or G.
  • the support is a microplate.
  • Electrophoresis is the separation of ionic molecules such as polypeptides by differential migration patterns through a gel based on the size and ionic charge of the molecules in an electric field. Electrophoresis can be conducted in a gel, capillary or on a chip. Examples of gels used for electrophoresis include starch, acrylamide, agarose or combinations thereof. A gel can be modified by its cross-linking, addition of detergents, immobilization of enzymes or antibodies (affinity electrophoresis) or substrates (zymography) and pH gradient. Methods to obtain polypeptides from electrophoresis gels are known to those of ordinary skill in the art.
  • CE capillary electrophoresis
  • CE may be used for separating complex hydrophilic molecules and highly charged solutes.
  • Advantages of CE include its use of small samples (sizes ranging from 0.001 to 10 ⁇ L), fast separation, easy reproducibility, very high efficiencies, meaning hundreds of components can be separated at the same time, is easily automated, can be used quantitatively and consumes limited amounts of reagents.
  • CE technology in general, relates to separation techniques that use narrow bore fused-silica capillaries to separate a complex array of large and small molecules.
  • CE can be further segmented into separation techniques such as capillary zone electrophoresis (CZE), capillary isoelectric focusing (CIEF) and capillary electrochromatography (CEC).
  • CZE capillary zone electrophoresis
  • CIEF capillary isoelectric focusing
  • CEC capillary electrochromatography
  • Capillary zone electrophoresis also known as free-solution CE (FSCE)
  • FSCE free-solution CE
  • the separation mechanism of CZE is based on differences in the charge-to-mass ratio of the analytes. Fundamental to CZE are homogeneity of the buffer solution and constant field strength throughout the length of the capillary. The separation relies principally on the pH-controlled dissociation of acidic groups on the solute or the protonation of basic functions on the solute.
  • Capillary isoelectric focusing allows amphoteric molecules, such as polypeptides, to be separated by electrophoresis in a pH gradient generated between the cathode and anode. A solute will migrate to a point where its net charge is zero. At this isoelectric point (the solute's pI), migration stops and the sample is focused into a tight zone. In CIEF, once a solute has focused at its pI, the zone is mobilized past the detector by either pressure or chemical means.
  • CEC is a hybrid technique between traditional liquid chromatography (HPLC) and CE.
  • HPLC liquid chromatography
  • CE capillaries are packed with HPLC packing and a voltage is applied across the packed capillary, which generates an electro-osmotic flow (EOF).
  • EEF electro-osmotic flow
  • the EOF transports solutes along the capillary towards a detector. Both differential partitioning and electrophoretic migration of the solutes occurs during their transportation towards the detector, which leads to CEC separations. It is therefore possible to obtain unique separation selectivities using CEC compared to both HPLC and CE.
  • the beneficial flow profile of EOF reduces flow related band broadening and separation efficiencies of several hundred thousand plates per meter are often obtained in CEC.
  • CEC also makes it is possible to use small-diameter packings and achieve very high efficiencies.
  • Micellar electrokinetic capillary chromatography is a capillary electropheretic method that allows the separation of uncharged solutes.
  • surfactants such as sodium dodecyl sulfate
  • the surface of anionic micelles of this type has a large negative charge, which give them a large electrophoretic mobility toward the positive electrode.
  • Most buffers exhibit such a high electroosmotic rate toward the negative electrode that the anionic micelles are carried toward the negative electrode, but at a much reduced rate. This form a fast moving aqueous phase and a slower moving micellar phase.
  • the components distribute themselves between the aqueous phase and the hydrocarbon phase at the interior of the micelles.
  • ITP isotachophoresis
  • ITP isotachophoresis
  • two different buffer systems are used to create zones which the analytes separate into.
  • ITP a large volume of sample is placed between a leading electrolyte and a terminating electrolyte. Analytes in the sample stack into narrow bands one after another according to their mobility.
  • the technique can be used in conjunction with capillary electrophoresis where a discontinuous electrolyte system is employed at the site of sample injection into the capillary.
  • transient isotachophoresis is a variation of this technique commonly used in conjunction with capillary electrophoresis (CE).
  • CE capillary electrophoresis
  • One configuration employs two reservoirs connected by a capillary.
  • the capillary and one reservoir are filled with a leading electrolyte (LE), while the second reservoir is filled with terminating electrolyte (TE).
  • LE leading electrolyte
  • TE terminating electrolyte
  • the sample for analysis is first injected into the capillary filled with LE and the injection end of the capillary is inserted into the reservoir containing TE. Voltage is applied and those components of the sample which have mobilities intermediate to those of the LE and TE stack into sharp ITP zones and achieve a steady state concentration. The concentration of such zones is related to the concentration of the LE co-ion but not to the concentration of the TE.
  • the reservoir containing TE is replaced with an LE containing reservoir. This causes a destacking of the sharp ITP zones, which allows individual species to move in a zone electrophoretic mode.
  • BGE background electrolyte
  • the leading zone will broaden due to electromigration dispersion and the concentration of higher mobility salt will decrease. The result is decreasing differences of the electric field along the migrating zones.
  • the sample bands will destack and move with independent velocities in a zone electrophoretic mode. Isolation of peptides can involve any procedure known in the art, such as capillary electrophoresis (e.g., in capillary or on-chip), or chromatography (e.g., in capillary, column or on a chip).
  • the contaminants can be inhibitors, interfering substances or inappropriate buffers.
  • removal of contaminants will be achieved by specifically purifying their protein of interest away from a complex mixture of biological molecules.
  • removal of contaminants will be achieved by specifically removing contaminants from a sample containing a protein of interest.
  • immobilized Protein A can be used to selectively remove immunoglobulins from a sample where they are considered to be a contaminant.
  • filters can be used to remove undesired components from a sample. Examples include but are not limited to size exclusion chromatography and ultrafiltration membranes that separate molecules on the basis of size and molecular weight.
  • ultracentrifugation is used for removing undesired components from a sample. Ultracentrifugation can involve centrifugation of a sample while monitoring with an optical system the sedimentation (or lack thereof) of particles.
  • electrodialysis is used to remove undesired components from the sample. Electrodialysis is an electromembrane process in which ions are transported through ion permeable membranes from one solution to another under the influence of a potential gradient. Since the membranes used in electrodialysis have the ability to selectively transport ions having positive or negative charge and reject ions of the opposite charge, electrodialysis is useful for concentration, removal, or separation of electrolytes.
  • Endotoxins are pyrogenic lipopolysaccharide (LPS) components of Gram-negative bacteria. Because these bacteria are ubiquitous, it is not surprising that endotoxins are frequent contaminants of biochemical preparations. Endotoxin contamination usually is measured as endotoxin units (EU), where 1 EU corresponds to a concentration of endotoxin (usually about 0.1 ng/kg body weight) sufficient to generate a pyrogenic reaction. In one instance removal of endotoxin is performed by ultracentrifugation. In another instance removal of endotoxin is performed by using immobilized polymixin B.
  • EU endotoxin units
  • Methods for reducing endotoxin levels include, but are not limited to, purification techniques using silica supports, glass powder or hydroxyapatite, reverse-phase, affinity, size-exclusion, anion-exchange chromatography, hydrophobic interaction chromatography, a combination of these methods, and the like.
  • Methods for measuring endotoxin levels include, but are not limited to, Limulus Amebocyte Lysate (LAL) assays.
  • LAL Limulus Amebocyte Lysate
  • detergent removal can occur by dialysis. Dialysis is effective for removal of detergents that have high CMCs (critical micelle concentrations) and/or small aggregation numbers, such as the N-octyl glucosides. In another instance removal of detergent from the sample can occur by sucrose density gradient separation. In yet another instance, detergents can be removed from the sample by size exclusion chromatography.
  • isolation of polypeptides may use genetic engineering techniques to synthesize of hybrid proteins.
  • a hybrid protein with an affinity tag can be produced directly by a microorganism. Examples of expression systems are Escherichia coli, Bacillus subtilis, Pseudomonas fluorescens, Pseudomonas aeruginosa, Pseudomonas putida, yeast, mammalian cells and the baculovirus system in insect cells.
  • the affinity tag can then be used to recover the product from a culture medium, cell lysate, estract, inclusion bodies, periplasmic space of the host cells, cytoplasm of the host cells, or other material by affinity chromatography.
  • non-natural amino acid polypeptides which are secreted into the medium can be obtained by centrifugation or filtration. These solutions may be suitable for direct application to chromatography columns.
  • polypeptides which are accumulated intracellularly are extracted prior to purification by chromatography.
  • polypeptides are extracted by cell disruption. Examples of cell disruption techniques include mechanical desintegrators, such as glass bead mills and high-pressure homoganizers.
  • polypeptides are extracted by cell permeabilization. Examples of permeabilization agents include but are not limited to guanidine hydrochloride and Triton X-100.
  • permeabilization agents include but are not limited to guanidine hydrochloride and Triton X-100.
  • chemical permeabilization cells can be permeabilized by enzymatic lysis. The clarification of the cell homogenate or crude extract obtained after cell permeabilization can be done by centrifugation or by different filtration methods, such as microfiltration or ultrafiltration.
  • hybrid polypeptides with a polyarginine tag can be purified by ion exchange chromatography
  • hybrid peptides with a polyphenylalanine tag can be isolated by hydrophobic chromatography
  • hybrid peptides with a ⁇ - Galactosidase tag can be isolated by affinity chromatography
  • hybrid peptides with a protein A tag can be isolated by IgG- affinity chromatography
  • hybrid peptides with an antigenic tag can be isolated by immunoaffinity chromatography
  • hybrid peptides with a polyhistidine can be isolated by metal chelate chromatography.
  • Tags may be removed by chemical or enzymatic means. In some embodiments, the tag is removed via an intramolecular reaction. A linker molecule may or may not be released.
  • non-natural amino acids may be used to generate purification tags and hybrid polypeptides with these tags can be purified using chromatography or other techniques.
  • multiple non-natural amino acids are included at a terminus of the polypeptide. Purification of this polypeptide with multiple non-natural amino acids may be purified by affinity chromatography or by other means depending on the properties of the non-natural amino acids.
  • the following procedure may be performed. After the binding of the polypeptide to a resin that binds to the non-natural amino acid tag, a reaction is performed to conjugate the polypeptide to another molecule such as PEG. The conjugated product may be released from the resin as a result of the conjugation or after the conjugation is complete. The conjugation may be performed under denaturing conditions and refolding of the polypeptide may be performed on the resin.
  • the second molecule may be conjugated to the polypeptide at a natural or non-natural amino acid present in the polypeptide.
  • the second molecule may be conjugated to the polypeptide at a natural or non-natural amino acid present in the non-natural amino acid tag.
  • the multiple non-natural amino acids included at a terminus of the polypeptide are metal-binding amino acids. Purification of this polypeptide may be performed using methods similar to those used for His-tagged proteins.
  • the polypeptide comprises two or more non-natural amino acids in that one or more non-natural amino acid is used to bind the polypeptide to a resin and the second non-natural amino acid is used to conjugate the polypeptide to another molecule, including but not limited to, PEG. Other materials useful in purification techniques may be used instead of resins.
  • Tags may be removed by chemical or enzymatic means. In some embodiments, the tag is removed via an intramolecular reaction. A linker may or may not be released.
  • a hybrid polypeptide may have a non-natural amino acid at the junction of the polypeptide and the tag.
  • This non-natural amino acid may be used to separate the polypeptide from the tag by chemical cleavage, for example during or after the binding of the tag to a column.
  • This non-natural amino acid may be used to separate the polypeptide from the tag by enzymatic cleavage or by an intramolecular chemical reaction.
  • a "prodrug” type approach is used.
  • a non-natural amino acid polypeptide is bound to a purification matrix, and a portion or all of the polypeptide is released after an event, including but not to, an intramolecular reaction, exposure to UV light (light activated molecule for release), chemical cleavage, or enzymatic cleavage.
  • a specific cleavage site at the junction between parts of a polypeptide could be introduced. This enables, for example, cleavage of the hybrid molecule to yield the protein of interest free of an affinity tag.
  • Removal of a fusion sequence may be accomplished by enzymatic or chemical cleavage.
  • a specific chemical or enzymatic cleavage site may be engineered into the fusion proteins. Enzymatic removal of fusion sequences may be accomplished using methods known to those of ordinary skill in the art. The choice of enzyme for removal of the fusion sequence will be determined by the identity of the fusion, and the reaction conditions will be specified by the choice of enzyme as will be apparent to one of ordinary skill in the art.
  • Chemical cleavage may be accomplished using reagents known to those of ordinary skill in the art, including but not limited to, cyanogen bromide, TEV protease, and other reagents.
  • cleavage reagents include but are not limited to, formic acid, hydroxylamine, collagenase, factor Xa, enterokinase, renin, carboxypeptidase A and carboxypeptidase B.
  • the cleaved hGH polypeptide may be purified from the cleaved fusion sequence and cleavage reagents by methods known to those of ordinary skill in the art.
  • Methods for purification may include, but are not limited to, size-exclusion chromatography, hydrophobic interaction chromatography, ion-exchange chromatography or dialysis or any combination thereof.
  • FIG. 10 shows an example of a purification method for a non-natural amino acid polypeptide utilizing a resin that reacts with the non-natural amino acid.
  • a covalent linkage is formed between a chemically specific affinity tag on the resin and a non-natural amino acid present in the protein. Such linkages are stable under a broad range of pH and purification conditions.
  • the separation step may be performed in alternate modes, including but not limited to a bath mode, enabling the large-scale purifications.
  • the resin and the affinity tags are physically and chemically stable, and thus, can be reused to reduce the cost of protein purification upon scale-up.
  • the separation can be performed in conjunction with conjugation of the polypeptide to molecules including but not limited to, PEG.
  • This "one-pot" method further simplifies the conjugation process and reduces the cost of production of proteins, including but not limited to target therapeutic proteins ( Figure 11 ).
  • Resins can be selected and functionalized according to the non-natural amino acid present in the polypeptide.
  • Figure 12 shows an example of resin selection and functionalization.
  • Resins or other matrixes for purification can be functionalized with different functional groups depending on the non-natural amino acid in the polypeptide. For example, FIG.
  • FIG. 13 shows an example of affinity purification of a non-natural amino acid polypeptide using hydroxylamine resin.
  • FIG. 14 shows an example of purification of a non-natural amino acid polypeptide using an aldehyde resin. The ability to regenerate the matrix used in purification methods also provides advantages for large-scale production.
  • the purification process changes one or more non-natural amino acids present in the polypeptide to one or more natural amino acids.
  • FIG. 15 shows an example of purification of native proteins from a non-natural amino acid precursor. The non-natural amino acid is converted to tyrosine after release from the resin used in the purification process.
  • FIG. 16 shows non-limiting examples of non-natural amino acids.
  • Non-natural amino acids present in a set of two or more proteins may be used to purify complexes of polypeptides.
  • the non-natural amino acids may be bonded to each other or joined via a linker, a polymer, or another molecule to enable purification of a complex of polypeptides.
  • Polypeptides that may be isolated in this fashion include but are not limited to multiple subunit receptors or enzymes.
  • Techniques used to isolate complexes may utilize one or more additional non-natural amino acids present in one or more of the polypeptides. Techniques for isolating large proteins are known to one of ordinary skill in the art.
  • Dissociation of the polypeptide complex may be performed using one or more non-natural amino acids present in one or more of the polypeptides.
  • One or more of the non-natural amino acids may be reacted with another molecule with a functional group that causes separation of the polypeptides in the complex.
  • the polypeptides may form a complex due to non-covalent interactions that involve one or more non-natural amino acids present in the polypeptide.
  • electro/chemical interaction such as electrical or magnetic fields may be used to purify polypeptides due to one or more non-natural amino acids present in the polypeptide.
  • single cell purification or isolation may be achieved using non-natural amino acid polypeptide.
  • the technological approaches for the screening process of the non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof disclosed herein include, but not limited to, multiwell-plate based screening systems, cell-based screening systems, microfluidics-based screening systems, and screening of soluble targets against solid-phase synthesized drug components.
  • Automated multiwell formats are developed high-throughput screening systems.
  • Automated 96-well plate-based screening systems are widely used.
  • the plate based screening systems can be made to reduce the volume of the reaction wells further, thereby increasing the density of the wells per plate.
  • Other types of high-throughput assays such as miniaturized cell-based assays can also be used in the present invention.
  • Miniaturized cell-based assays have the potential to generate screening data of quality and accuracy, due to their in vivo nature.
  • Microfluidics-based screening systems that measure in vitro reactions in solution make use of ten to several-hundred micrometer wide channels. Micropumps, electroosmotic flow, integrated valves and mixing devices control liquid movement through the channel network.
  • Libraries for screening can be grouped as, by way of example only, General Screening or Template-Based such as Groups with common heterocyclic lattices; Targeted such as Mechanism based selections, for example, Kinase Modulators, GPCR Ligands, Antiinfectives, Potassium Channel Modulators, and Protease Inhibitors; Privileged Structure such as Compounds containing chemical motifs that are more frequently associated with higher biological activity than other structures; Diversity such as Compounds pre-selected from available stock with maximum chemical diversity; Plant Extracts; Natural Products / Natural Product-Derived, etc.
  • Mechanism based selections for example, Kinase Modulators, GPCR Ligands, Antiinfectives, Potassium Channel Modulators, and Protease Inhibitors
  • Privileged Structure such as Compounds containing chemical motifs that are more frequently associated with higher biological activity than other structures
  • Diversity such as Compounds pre-selected from available stock with maximum chemical diversity
  • Plant Extracts Natural Products / Natural Product-
  • Combinatorial chemical libraries are a means to assist in the generation of new chemical compound leads.
  • a combinatorial chemical library is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical "building blocks” such as reagents. Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks.
  • LogP, molecular weight, number of H-bond donors and acceptors, as set forth in the Lipinski "rule of five" requirements help to determine strong candidates for drug-like characteristics. Lipinski "rule of five” requires the compound to have these properties: five or fewer hydrogen bond donors, molecular weight less than or equal to 500 Da, calculated LogP less than or equal to 5), and ten or fewer hydrogen bonding acceptors.
  • High throughput screening technologies coupled with compound libraries obtained through combinatorial chemistry and/or high throughput synthesis methods can be utilized to rapidly identify and optimize ligands for non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof, as disclosed herein.
  • Chemical diversity libraries of organic compounds include, but are not limited to: benzodiazepines, diversomers such as hydantoins, benzodiazepines and, analogous organic syntheses of small compound libraries, oligomeric libraries such as peptide, N-alkyl glycine, polycarbamate and polyureas, oligocarbamates, and/or peptidyl phosphonates, carbohydrate libraries, chiral compound libraries, and small organic molecule libraries.
  • oligomeric libraries such as peptide, N-alkyl glycine, polycarbamate and polyureas, oligocarbamates, and/or peptidyl phosphonates
  • carbohydrate libraries chiral compound libraries
  • small organic molecule libraries A wide variety of heterocyclic compound libraries have been synthesized by solid phase methods.
  • benzodiazepins include, by way of example only, benzodiazepins, pyrrolidines, hydantoins, 1,4-dihydropyridines, isoquinolinones, diketopiperazines, benzylpiperazines, quinolones, dihydro- and tetrahydroisoquinolines, 4-thiazolidinones, b-lactams, benzisothiazolones, pyrroles and imidazoles.
  • Combinatorial libraries of inorganic compounds include, but not limited to, (a) Oxides of metals and main group elements, including transition metal oxides such as zirconia, titania, manganese oxide, rare earth oxides such as ceria and lanthanum oxide; binary, ternary, and more complex solid state oxides and ceramic phases; various forms of alumina, silica, aluminosilicates and aluminophosphates; (b) Natural and synthetic forms of aluminosilicate and silicate zeolites such as ZSM-5, Beta, zeolite Y, and ferrierite, various forms of molecular sieves such as aluminophosphates and titanosilicates; natural or synthetic clays and related minerals such as kaolin, attapulgite, talc, montmorillonite, and Laponite®; (c) Non-oxide ceramics such as metal carbides and nitrides; (d) Various forms of carbons such as activated carbon,
  • Peptide library by using microorganisms - Antibodies and immune cell receptors of the immune system are representative biological libraries.
  • all the processes of library design, synthesis, and optimization are controlled by the organism itself. Only structures of antigens and genetic information to form embryonic factors are external conditions, but the rest is controlled spontaneously by internal factors.
  • the immune system uses protein structure libraries, they are libraries using amino acids as basic factors. Because peptides or proteins made of amino acids are the first products of synthesis by translating genetic information, through genetic engineering technologies, proteins of desired sequences can be easily obtained by inserting modified genetic information into microorganisms like bacteria or virus. Microorganism library synthesis brings several advantages.
  • microorganisms It is possible to clone microorganisms to make only one kind of proteins per microorganism, and even though only one cell is acquired, the number of clones can be easily increased by cell multiplication.
  • the other advantage of using microorganisms is that they can self-propagate whenever there is enough supply. After synthesizing a DNA strand that makes the desired protein sequence, its complementary strand is synthesized, by enzymes if needed. For synthesized DNA to replicate and translate properly in microorganisms, it needs to be packed with vector and inserted into microorganisms. Proteins expressed on the surface of the microorganism, and to find desired proteins is the next step.
  • Random DNA synthesis or cutting cDNA or the whole genomic DNA of a particular organism can be used.
  • a portion of DNA sequence that makes particular protein can be modified to make mutated protein library.
  • 10 9 (one billion) kinds of libraries can be made.
  • the number of 5-unit peptides is 20 5 (3.2 millions), that of 6-unit ones is 64 millions, and for 7-unit peptides the number passes one billion. Therefore, if more than 7 amino acids are changed incomplete library that does not contain all the possible combinations is made. For long proteins, 7 different amino acids can be selected separately and replaced.
  • DNA codes can be repeated and designate the same amino acid, and generation frequency changes. Therefore, to make all the possible combinations, much more quantities of clones are required.
  • a linear combinatorial biological library such as a polypeptide library is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound).
  • the proteins may be members of a protein family such as a receptor family (examples: growth factor receptors, catecholamine receptors, amino acid derivative receptors, cytokine receptors, lectins), ligand family (examples: cytokines, serpins), enzyme family (examples: proteases, kinases, phosphatases, ras-like GTPases, hydrolases), transcription factors (examples: steroid hormone receptors, heat-shock transcription factors, zinc-finger, leucine-zipper, homeodomain), HIV proteases or hepatitis C virus (HCV) proteases, and antibody or antibody fragment (Fab, for example).
  • a receptor family examples: growth factor receptors, catecholamine receptor
  • peptoids encoded peptides
  • random biooligomers dipeptides
  • vinylogous polypeptides vinylogous polypeptides
  • nonpeptidal peptidomimetics with Beta D Glucose scaffolding antibody libraries
  • peptide nucleic acid libraries peptide nucleic acid libraries
  • Bacteriophage library It is one of a number of protein library methods. Bacteriophage is living in a host bacterium and a kind of virus with genetic materials and capsids. M 13 and Lambda viruses are the most famous.
  • a M13 is a thin, long virus and due to its small genome size, numerous libraries can be made easily. Different from other viruses, it can come out to outside of host cells without damaging them or inhibiting their growth. It is known that M13 amplifies its genetic information in the host cell and wears the capsid when emerging. It makes 10 kinds of proteins and pVIII and pIII capsids are commonly used in library synthesis among them.
  • a pVIII protein surrounds the whole body and has about 50 amino acids. Usually 2700 per a virus are expressed. Because its amino end protrudes toward outside of the capsid, it can be modified to express a different peptide on it. Usually a long peptide cannot be expressed, but it is possible for 6-unit peptides.
  • a pIII protein is expressed at the end of a virus, and usually 3 to 5 proteins of 406 amino acids are expressed. It can express quite large proteins so that it is used for the whole protein or antibody molecule libraries.
  • a normal antibody uses Fab, an antigen recognition region, or a Fvs chain.
  • Bacteriophage Library and hybridoma are the most famous methods to make antibodies. M13 is ideal to make random peptide libraries and the virus is stable enough to be precipitated and concentrated so that screening 10 9 libraries in a volume of 1 - 10 ⁇ L is possible.
  • a Lambda virus coats itself with a capsid in the cytoplasm and comes out of its host cell when there is an enough number, instead of wearing a capsid when emerging. In other words, if a different protein is expressed, it will probably emerge in a folded shape with proper functions.
  • a pV and D proteins are commonly used for the library synthesis. As proteins that can be expressed on a bacteriophage surface, there are random peptide, natural protein fragments, mutated particular protein libraries, and partial antibody fragments and they are used for chromatography materials, protein-protein mutual reactions, receptor binding site searching, and drug discoveries.
  • Phage display is a widely utilized technique to make peptide libraries. These peptide libraries are useful for screening to identify peptides that have a particular desired activity, such as binding to another polypeptide or other molecule.
  • phage display the peptide library is fused to a bacteriophage protein, typically a coat protein,that is displayed on the surface of the phage.
  • the library of peptide bearing phage is contacted with an immobilized binding partner, such as a cell surface or a purified protein, and specific binders are then isolated.
  • Phage display techniques and libraries are described in US Patents No.
  • U.S. Patent No. 5,750,373 describes a method for selecting novel proteins such as growth hormone and antibody fragment variants having altered binding properties for their respective receptor molecules. The method comprises fusing a gene encoding a protein of interest to the carboxy terminal domain of the gene III coat protein of the filamentous phage M13.
  • Bacteria and yeast libraries Not only viruses with capsids, but also bacteria with cell walls and membranes can be used for library expression as well. Both the gram-positive bacteria and gram-negative bacteria can be used to express proteins on cell surfaces, and E. coli, a gram-negative bacterium, is commonly used. Bacteria library can find an antigen that strongly binds to a certain antibody and use it as a vaccine, or it can express diagnostic antibodies or receptor libraries for analysis of particular materials.
  • FACS fluorescence-activated cell sorting
  • Fluorescence labeled target molecules are added to the library of proteins expressed on a cell surface and flow through thin tubes of FACS machine.
  • FACS sorts each cells by fluorescent colors and intensities as alive. It is possible to screen different target molecules with different colors and also possible to sort cells of different intensities and selectivity. Another advantage is a liquid-phase screening. It is not necessary to separate strongly clung molecules. Sorted cells multiply again and they are re-screened.
  • Yeast surface display techniques are also widely utilized to product and display peptide libraries. Yeast surface display may be utilized in combination with fluorescence activated cell sorting to select cells displaying the desired peptides. Yeast surface display techniques and libraries are described in US Patents No. 6083693 , 6406863 , 6410271 , 6232074 , 6410246 , 6610472 .
  • Bacterial surface display has been used in a variety of forms to display peptides on the cell surface or in the periplasm.
  • a variety of bacterial hosts are available for use in this system, as are a variety of polypeptide anchoring domains to anchor the displayed peptide to the cell surface.
  • Bacterial surface display techniques and libraries are described in US Patents No. 5348867 , 5866344 , 6277588 , 5635182 , 6180341 .
  • in vivo systems are utilized to make libraries of polypeptides and identify changes in activities, such as target protein binding modulation, resulting from changes in amino acid sequences.
  • in vivo systems include, but are not limited to, the yeast two hybrid system ( Schneider, S et al., Nat. Biotechnol., 17, 170-175 (1990 )), and the dihydrofolate reductase protein-fragment complementation assay ( Pellitier, N.J. et al., Nat. Biotechnol., 17, 683-690, (1990 )).
  • Bio-panning - A synthesized microorganism library may be used to find a peptide that binds to a particular molecule with high affinity.
  • Target molecules such as non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof as disclosed herein, may be evenly placed on a test plate.
  • the prepared microorganism library may be added to the plate. Only the microorganisms that strongly bind to the target molecules will remain and the rest will be in the solution. After a while, unbound microorganisms may be discarded, and then weakly or accidentally bound microorganisms may be washed with appropriate solutions.
  • the target molecule's binding affinity determines the washing process. Still remaining microorganisms can be taken apart by addition of low pH or high concentrated target molecules, and the quantity is amplified by re-incubation.
  • each clone may be separated and usually tens of clones may be selected and used for DNA sequence analysis. It is successful if peptide structures from DNA information are recognizable and most of clones show accord peptide sequences.
  • the microorganism protein library technology fundamentally uses a living organism's self-reproduction ability. That is, by amplifying (feeding) a small quantity of obtained candidate molecules, one can increase purity and quantity.
  • Ribosome display and mRNA display techniques are also widely utilized to make peptide libraries.
  • Ribosome display and mRNA display are in vitro techniques that couple the mRNA encoding a peptide to the encoded peptide either on the ribosome or by using puromycin.
  • Ribosome display and mRNA display techniques and libraries are described in US Patents No.
  • 5' end and 3' end are fixed in a sequence and A, T, C, and G are randomly placed as each take about 25% of the sequence.
  • one strand is made, it may be replicated by using enzymes or amplified by PCR. Commonly about 1014-15 molecules are made and used, but occasionaly there are about 40 places (1024 kinds) for random introduction, sometimes they start with incomplete set of library.
  • DNA DNA themselves are simply used, but for RNA library, T7 RNA Polymerase is needed to transcript.
  • RNA RNA-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding-binding and amplification of the amplified library is repeated until the beginning number of 10 14-15 is narrowed to several hundreds, and then sequences of acquired candidate molecules are analyzed and each binding affinity is measured. Such acquired DNA and RNA are called aptamers, and they show strong affinity toward protein target molecules. The aptamer inhibits the target molecule's function in vivo, but it is quickly destroyed by in vivo nucleases. To solve the problem, some parts of library are substituted with artificial nucleic aids to increase resistance against nucleases.
  • Bioactive Lipid Library Endocannabinoid Library- compounds having activity at cannabinoid (CB) and vanniloid (VR) receptors which includes various classes of ligands, for example, Amides, Ethanolamides, Lipo-amino acids, Acyl-GABAs, and Acyl-dopamines etc.
  • Known Bioactives Library such as, GPCR ligands, second messenger modulators, nuclear receptor ligands, actin & tubulin modulators, kinase inhibitors, protease inhibitors, ion channel blockers, gene regulation agents, lipid biosynthesis inhibitors, etc.
  • Ion Channel Ligand Library Kinase/Phosphatase Inhibitor Library
  • Natural Products Library- Natural products are an unsurpassed source of chemical diversity and are an ideal starting point for any screening program for pharmacologically active small molecules
  • Neurotransmitter Library- CNS Receptor Ligands such as, Ad
  • Orphan Ligand Library-Orphan ligand library contains compounds with biological activity but whose protein binding partners have not been identified. For example, trace Amines, neurotransmitter metabolites, endogenouse ß-carbolines, urinary metabolites, nicotine congeners, and D-Amino Acids etc.
  • Assays may be conducting in a variety of ways including screening a library of non-natural amino acid polypeptides with a known molecule or vice versa.
  • the method is performed in single test tubes or on a modest scale.
  • the method is performed in plurality simultaneously.
  • the method can be performed at the same time on multiple assay mixtures in a multi-well screening plate.
  • fluorescence or absorbance readouts are utilized to determine activity.
  • Other methods may also include protein chip systems which can screen enzymes, receptor proteins or antibodies which aid conducting protein-protein interaction studies, ligand binding sutdies, or immunoassays ( MacBeath and Schreiber, Science 2000 289: 1760-1763 ).
  • Another instance may involve, profiling drug which can effect in intact cells, that are introduced with functional non-natural amino acid polypeptides, by probing the cell physiology using fluorescent stains for DNA and other proteins known to interact with the non-natural amino acid polypeptide and using fluorescent microscopes generated pictures so as to measure changes in the cells' behavior ( Mayer, T.U., Kapoor, T.M., Haggarty, S.J., King, R.W., Schreiber, S.L., Mitchison, T.J. (1999). Science.286, 971-4 .)
  • the detection method is carried out on a test combination (test ligand-non-natural amino acid polypeptide combination) after sufficient time has passed for binding of a non-natural amino acid polypeptide to its ligand and on a control combination (which is the same as the test combination except that no test ligand is present).
  • a test ligand may be combined with a non-natural amino acid polypeptide for which a ligand ( i . e ., an agent which binds the non-natural amino acid polypeptide) is to be identified.
  • the resulting combination is a test ligand- non-natural amino acid polypeptide combination or test combination.
  • the test ligand is present in excess molar amounts, relative to the non-natural amino acid polypeptide.
  • the present method can be carried out in solution or, in some embodiments of the method, the non-natural amino acid polypeptide can be present on a solid phase ( e . g ., linked covalently through a linker or otherwise to a bead).
  • test ligand and non-natural amino acid polypeptide are combined under conditions (e . g ., temperature, pH, salt concentration, time) appropriate for binding of the non-natural amino acid polypeptide to a ligand.
  • conditions under which test ligand and non-natural amino acid polypeptide are combined are generally such that, for non-natural amino acid polypeptide that unfolds reversibly, a substantial fraction of non-natural amino acid polypeptide is present in the absence of the test ligand in the unfolded form, although the fraction can vary, depending on the detection method used.
  • conditions are generally such that the non-natural amino acid polypeptide unfolds at a substantial rate in the absence of ligand.
  • the conditions under which the present method is carried out will be determined empirically, using known methods. Such conditions include reaction temperature and the chaotropic agent(s) or denaturant(s) used. The temperature at which the method is carried out is determined by the non-natural amino acid polypeptide being used and can be determined empirically using known methods.
  • denaturing conditions may be required for some non-natural amino acid polypeptide.
  • denaturing conditions might include the use of elevated temperatures, the addition of protein denaturants (e.g., urea, guanidine) to the incubation mixture or use of both.
  • protein denaturants e.g., urea, guanidine
  • stability of some non-natural amino acid polypeptide might be adjusted through engineering destabilizing or stabilizing amino acid substitutions in the non-natural amino acid polypeptide.
  • the test ligand and non-natural amino acid polypeptide are combined, maintained under appropriate conditions and for sufficient time for binding of the non-natural amino acid polypeptide to a ligand.
  • the time necessary for binding of non-natural amino acid polypeptide to ligand will vary depending on the test ligand, non-natural amino acid polypeptide and other conditions used. In some cases, binding will occur instantaneously ( e.g., essentially simultaneous with combination of test ligand and non-natural amino acid polypeptide), while in others, the resulting test ligand- non-natural amino acid polypeptide combination is maintained for a longer time before binding is detected. In the case of non-natural amino acid polypeptide which unfolds irreversibly, the rate of unfolding must also be taken into consideration in determining an appropriate time for binding of test ligand.
  • Binding of a test ligand to the non-natural amino acid polypeptide is assessed in one of several ways: by determining the extent to which folded non-natural amino acid polypeptide is present in the test ligand- non-natural amino acid polypeptide combination; by determining the extent to which unfolded non-natural amino acid polypeptide is present in the test ligand- non-natural amino acid polypeptide combination or by determining the ratio of folded non-natural amino acid polypeptide to unfolded non-natural amino acid polypeptide in the combination.
  • the difference between the amount of folded non-natural amino acid polypeptide, the amount of unfolded non-natural amino acid polypeptide or the ratio of folded non-natural amino acid polypeptide to unfolded non-natural amino acid polypeptide in the presence of the test ligand and in its absence is determined.
  • test ligand binds the non-natural amino acid polypeptide (i.e., if the test ligand is a ligand for the non-natural amino acid polypeptide), there will be more folded non-natural amino acid polypeptide and less unfolded non-natural amino acid polypeptide (and, thus, a higher ratio of folded to unfolded non-natural amino acid polypeptide and a lower ratio of unfolded to folded non-natural amino acid polypeptide) than is present in the absence of a test ligand which binds the non-natural amino acid polypeptide. It is not necessary to determine the quantity or fraction of a folded and unfolded non-natural amino acid polypeptide.
  • the difference between the extent to which the two forms occur in the absence of a test ligand can be assessed by determining their occurrence initially (e.g., prior to addition of a test ligand to a solution of non-natural amino acid polypeptide or to solid support-bound test protein) and then after the test ligand has been combined with the non-natural amino acid polypeptide under conditions appropriate for non-natural amino acid polypeptide -ligand binding to occur.
  • determination of the two forms of non-natural amino acid polypeptide can be carried out using a variety of known methods, which are described below.
  • a test ligand which is shown by the present method to bind a non-natural amino acid polypeptide is referred to as a ligand of the non-natural amino acid polypeptide.
  • binding of test ligand to non-natural amino acid polypeptide is detected through the use of proteolysis.
  • a protease which acts preferentially upon unfolded non-natural amino acid polypeptide is combined with the test ligand-non-natural amino acid polypeptide combination (test combination) and the resulting test combination-protease mixture is assayed after an appropriate period of incubation, using one of the methods described in detail below, to determine the difference between intact or degraded non-natural amino acid polypeptide in the presence and in the absence of the test ligand.
  • An identical assay is performed on a test ligand-non-natural amino acid polypeptide combination and on a control combination and results of the two assays are compared.
  • test ligand More intact protein or less degraded protein in the test combination than in the control combination indicates that the test ligand has bound the non-natural amino acid polypeptide and, thus, indicates that the test ligand is a ligand of the non-natural amino acid polypeptide. Similarly, a higher ratio of intact non-natural amino acid polypeptide to degraded protein in the test combination than in the control indicates the test ligand is a ligand of the non-natural amino acid polypeptide.
  • proteases such as trypsin, chymotrypsin, V8 protease, elastase, carboxypeptidase, proteinase K, thermolysin and subtilisin, can be used in this embodiment. It is only necessary that the protease used be able to act upon (hydrolyze the peptide bonds of) the non-natural amino acid polypeptide used under the chosen incubation conditions and that this action be preferentially directed toward the unfolded form of the protein. To avoid interference by target ligands which directly inhibit the protease, more than one protease can be used simultaneously or in parallel assays.
  • the peptide substrate--the non-natural amino acid polypeptide--must have access to the enzyme active site of the chosen protease. Because the atoms in a folded protein molecule are tightly packed, the majority of the susceptible peptide bonds are sterically blocked from entering a protease active site when the protein is in the folded state. In the unfolded state, the peptide bonds are more exposed and are therefore relatively more susceptible to protease action.
  • test ligand which binds the folded non-natural amino acid polypeptide, stabilizing it in the protease-resistant form, changes the rate of proteolysis.
  • the test ligand by incubating the test ligand with the non-natural amino acid polypeptide, adding a protease to preferentially degrade the unfolded proteins, and then employing an assay to quantify the intact or the degraded non-natural amino acid polypeptide, it is possible to ascertain whether the test ligand bound the non-natural amino acid polypeptide and, thus, is a ligand of the non-natural amino acid polypeptide, indicating that it is potentially therapeutically useful.
  • the protease may be intrinsic to the unpurified or partially purified non-natural amino acid polypeptide sample.
  • the propensity of unfolded proteins to adhere to surfaces is utilized.
  • This embodiment relies on the fact that folded proteins are held in specific three dimensional arrangements and, thus, are not as likely as their unfolded counterparts to bind a surface. If a test ligand binds a non-natural amino acid polypeptide (i.e., is a ligand of the non-natural amino acid polypeptide), it will stabilize the folded form of the non-natural amino acid polypeptide.
  • the ability of a test ligand to bind a non-natural amino acid polypeptide can be determined by assessing the extent to which non-natural amino acid polypeptide is bound to an appropriate solid surface in the presence and in the absence of the test ligand. The methods described in detail below can be used for this purpose.
  • the non-natural amino acid polypeptide, a test ligand and a surface that preferentially binds unfolded protein are combined and maintained under conditions appropriate for binding of the non-natural amino acid polypeptide to a ligand and binding of unfolded non-natural amino acid polypeptide to the surface.
  • suitable surfaces for this purpose including microtiter plates constructed from a variety of treated or untreated plastics, plates treated for tissue culture or for high protein binding, nitrocellulose filters and PVDF filters.
  • test ligand binds the non-natural amino acid polypeptide, more folded non-natural amino acid polypeptide and less unfolded non-natural amino acid polypeptide is present in the test ligand-non-natural amino acid polypeptide combination than is present in a comparable control combination. That is, in the presence of a test ligand that is a ligand for a non-natural amino acid polypeptide, less unfolded protein is available to bind a surface that preferentially binds unfolded protein than in the absence of a ligand for the non-natural amino acid polypeptide.
  • Determination of the amount of surface-bound non-natural amino acid polypeptide or the amount of non-natural amino acid polypeptide remaining in solution can be carried out using one of the methods described below. If more non-natural amino acid polypeptide is not surface bound (i.e., if more non-natural amino acid polypeptide is in solution) in the presence of a test ligand than in the absence of the test ligand, the test ligand is a ligand of the non-natural amino acid polypeptide.
  • the ratio of non-natural amino acid polypeptide in solution to surface-bound non-natural amino acid polypeptide is greater if a test ligand is a ligand for the non-natural amino acid polypeptide than if it is not. Conversely, the ratio of surface-bound non-natural amino acid polypeptide to non-natural amino acid polypeptide in solution is less if a test ligand is a ligand for the non-natural amino acid polypeptide than if it is not.
  • the extent to which folded and unfolded non-natural amino acid polypeptide are present and, thus, binding of test ligand to non-natural amino acid polypeptide are assessed through the use of specific antibodies directed against only the unfolded state ("denatured-specific antibodies” or "DS antibodies”) or only the folded state (“nature specific antibodies” or “NS antibodies”).
  • specific antibodies directed against only the unfolded state (“denatured-specific antibodies” or "DS antibodies” or only the folded state
  • DS antibodies specific antibodies directed against only the unfolded state
  • DS antibodies only the folded state
  • the DS antibody's apparent binding affinity will be reduced ( Breyer, (1989) "Production and Characterization of Mono-clonal Antibodies to the N-terminal Domain of the Lambda Repressor", J.
  • DS or NS antibodies can be utilized to detect a ligand-induced change in the occurrence of folded non-natural amino acid polypeptide, the occurrence of unfolded proteins or the ratio of one to the other.
  • a test solution containing the DS antibody directed against the unfolded non-natural amino acid polypeptide, the non-natural amino acid polypeptide, and the test ligand is incubated, such as in a microtiter plate coated with the denatured non-natural amino acid polypeptide or a peptide fragment thereof, under conditions appropriate for binding of the non-natural amino acid polypeptide with its ligand and binding of the DS antibody to unfolded non-natural amino acid polypeptide.
  • a control solution which is the same as the test solution except that it does not contain test ligand, is processed in the same manner as the test solution.
  • a test solution containing the DS antibody, the test ligand, and the non-natural amino acid polypeptide is incubated in a plate coated with a second antibody, referred to as a solid phase antibody, which cannot bind to the non-natural amino acid polypeptide simultaneously with the DS antibody, and is specific for the non-natural amino acid polypeptide, but is either specific for the folded state ("native specific” or "NS antibody”) or unable to differentiate between the native and denatured states (“non-differentiating" or "ND antibody”).
  • test combination or solution is maintained under conditions appropriate for binding of the non-natural amino acid polypeptide with a ligand of the non-natural amino acid polypeptide and for binding of the antibodies to the proteins they recognize (are specific for).
  • a control solution which is the same as the test solution except that it does not contain test ligand, is processed in the same manner as the test solution. In both solutions, denatured (unfolded) non-natural amino acid polypeptide binds the DS antibody and is inhibited from binding the solid phase antibody.
  • the ability of the test ligand to bind the non-natural amino acid polypeptide can be gauged by determining the amount of non-natural amino acid polypeptide that binds to the solid phase antibody in the test solution and comparing it with the extent to which non-natural amino acid polypeptide binds to the solid phase antibody in the absence of test ligand, which in turn reflects the amount of non-natural amino acid polypeptide in the folded state.
  • the amount of non-natural amino acid polypeptide bound to the plate via the second antibody or remaining in solution can be detected by the methods described below. This approach may be used in a comparable manner with NS antibody as the in solution antibody and DS or ND antibody on the solid phase.
  • a test solution containing the non-natural amino acid polypeptide and the test ligand is incubated in a container, such as a microtiter well which has been coated with a DS or NS antibody and maintained under conditions appropriate for binding of non-natural amino acid polypeptide to its ligand and for binding of the antibody to non-natural amino acid polypeptide.
  • the antibody can be present on the surfaces of beads. The ability of the test ligand to bind the non-natural amino acid polypeptide is gauged by determining the extent to which non-natural amino acid polypeptide remains in solution (unbound to the antibody) or on the solid surface (bound to the antibody), or the ratio of the two, in the presence and in the absence of test ligand.
  • test ligand binds the non-natural amino acid polypeptide (is a ligand of the non-natural amino acid polypeptide)
  • a ligand of the non-natural amino acid polypeptide there will be less non-natural amino acid polypeptide bound to a DS antibody or more bound to an NS antibody (i.e., more non-natural amino acid polypeptide will be in solution in the case of DS antibody or less in solution for NS antibody) than is bound to the antibody in the control solution.
  • the antibody can be present in solution and the non-natural amino acid polypeptide can be attached to a solid phase, such as a plate surface or bead surface.
  • molecular chaperones are used to determine binding of a test ligand to a non-natural amino acid polypeptide.
  • Chaperones are a variety of protein that bind unfolded proteins as part of their normal physiological function. They are generally involved in assembling oligomeric proteins, in ensuring that certain proteins fold correctly, in facilitating protein localization, and in preventing the formation of proteinaceous aggregates during physiological stress. Hardy, (1991) "A Kinetic Partitioning Model of Selective Binding of Nonnative Proteins by the Bacterial Chaperone SecB", Science 251:439-443 These proteins have the ability to interact with many unfolded or partially denatured proteins without specific recognition of defined sequence motifs.
  • SecB One molecular chaperone, found in E. coli, is SecB.
  • SecB has a demonstrated involvement in export of a subset of otherwise unrelated proteins. Competition experiments have shown that SecB binds tightly to all the unfolded proteins tested, including proteins outside of its particular export subset, but does not appear to interact with the folded protein.
  • a test solution containing the test ligand and the target is incubated on a microtiter plate or other suitable surface coated with molecular chaperones, under conditions appropriate for binding of non-natural amino acid polypeptide with its ligand and binding of the molecular chaperones used to unfolded non-natural amino acid polypeptide.
  • the unfolded non-natural amino acid polypeptide in the solution will have a greater tendency to bind to the molecular chaperone-covered surface relative to the ligand-stabilized folded non-natural amino acid polypeptide.
  • the ability of the test ligand to bind non-natural amino acid polypeptide can be determined by determining the amount of non-natural amino acid polypeptide remaining unbound, or the amount bound to the chaperone-coated surface, using the methods detailed below.
  • a competition assay for binding to molecular chaperones can be utilized.
  • a test solution containing purified non-natural amino acid polypeptide, the test ligand, and a molecular chaperone can be incubated in a container, such as a microtiter well coated with denatured (unfolded) non-natural amino acid polypeptide, under conditions appropriate for binding non-natural amino acid polypeptide with its ligand and binding of the molecular chaperones to unfolded non-natural amino acid polypeptide.
  • a control solution which is the same as the test solution except that it does not contain test ligand is processed in the same manner.
  • Denatured non-natural amino acid polypeptide in solution will bind to the chaperonin and, thus, inhibit its binding to the denatured non-natural amino acid polypeptide bound to the container surface (microtiter well surface). Binding of a test ligand to non-natural amino acid polypeptide will result in a smaller amount of unfolded non-natural amino acid polypeptide, and, thus, more chaperones will be available to bind to the solid-phase denatured non-natural amino acid polypeptide than is the case in the absence of binding of test ligand. Thus, binding of test ligand can be determined by assessing chaperones bound to the surface or in solution in the test solution and i D the control solution and comparing the results.
  • Binding of chaperone to solid-phase denatured non-natural amino acid polypeptide to a greater extent in the test solution than in the control solution is indicative of test ligand-non-natural amino acid polypeptide binding (i.e., is indicative of identification of a ligand of the non-natural amino acid polypeptide).
  • the molecular shaperones are generally not provided in excess, so that competition for their binding can be measured.
  • test solution containing the non-natural amino acid polypeptide, the test ligand and a molecular chaperone can be incubated in a container, such as a microtiter well, whose surface is coated with antisera or a monoclonal antibody specific for the folded non-natural amino acid polypeptide (NS antibody) and unable to bind the non-natural amino acid polypeptide bound to the chaperone.
  • a container such as a microtiter well, whose surface is coated with antisera or a monoclonal antibody specific for the folded non-natural amino acid polypeptide (NS antibody) and unable to bind the non-natural amino acid polypeptide bound to the chaperone.
  • Unfolded non-natural amino acid polypeptide will bind chaperone in solution and thus be inhibited from binding the solid phase antibody.
  • test ligand By detecting non-natural amino acid polypeptide in the solution or bound to the well walls and comparing the extent of either or both in an appropriate control (the same combination without the test ligand), the ability of the test ligand to bind non-natural amino acid polypeptide can be determined. If the test ligand is a ligand for the non-natural amino acid polypeptide, more non-natural amino acid polypeptide will be bound to the antisera or monoclonal antibody bound to the container surface in the test solution than in the control solution. Conversely, less non-natural amino acid polypeptide will be present unbound (in solution) in the test solution than in the control solution.
  • Detection and comparison of bound non-natural amino acid polypeptide, unbound non-natural amino acid polypeptide or a ratio of the two in the test solution and control solution indicate whether the test ligand is a ligand of the non-natural amino acid polypeptide or not.
  • the amount of non-natural amino acid polypeptide bound to the solid substrate can be assessed by sampling the solid substrate or by sampling the solution, using the detection methods outlined below.
  • unfolded protein For proteins that unfold irreversibly, unfolded protein often forms insoluble aggregates.
  • the extent of protein aggregation can be measured by techniques outlined below such as light scattering, centrifugation, and filtration. In this approach, non-natural amino acid polypeptide and test ligand are incubated and the amount of protein aggregation is measured over time or after a fixed incubation time. The extent of protein aggregation in the test mixture is compared to the same measurement for a control assay in the absence of test ligand. If a test ligand binds a non-natural amino acid polypeptide, the rate of unfolding of non-natural amino acid polypeptide will be lower than in the absence of test ligand.
  • the rate of increase of unfolded protein and hence of aggregated protein will be lower if the test ligand is a ligand for the non-natural amino acid polypeptide than if it is not.
  • the ability of a test ligand to bind a non-natural amino acid polypeptide can be determined by assessing the extent of protein aggregation in the presence and absence of test ligand.
  • Methods known in the art to detect the presence or absence of protein, small peptides or free amino acids can be used in the present method for detecting non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof
  • the method used can be determined by the product (proteins, peptides, free amino acids) to be detected.
  • techniques for detecting protein size can be used to determine the extent of proteolytic degradation of the non-natural amino acid polypeptide.
  • Radio-labeling, fluorescence labeling, and enzyme-linked labeling can detect the presence or absence either in solution or on a substrate by measurement of radioactivity, fluorescence or enzymatic activity.
  • Immunologic methods can detect the presence or absence of a known non-natural amino acid polypeptide in solution or on a substrate such as by binding of an antibody specific for that protein.
  • FIG. 1a presents various protein detection techniques that can be used to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • Methods for protein detection disclosed herein include fluorescence microscopy to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • Fluorescence Microscopy is a widely used microscopy technique that enables the molecular composition of the structures being observed to be identified through the use of fluorescently-labelled probes of high chemical specificity. Such probes may be antibodies, antibody fragments, or antigen-binding polypeptides that comprise a non-natural amino acid. Fluorescence microscopy may be used in studies of fixed specimens. For proteins that can be extracted and purified in reasonable abundance, a fluorophore may be conjugated to a protein and the conjugate introduced into a cell.
  • a fluorophore may be conjugated to a non-natural amino acid in the polypeptide. It is assumed that the fluorescent analogue behaves like the native protein and can therefore serve to reveal the distribution and behavior of this protein in the cell. Along with NMR, infrared spectroscopy, circular dichroism and other techniques, protein intrinsic fluorescence decay and its associated observation of fluorescence anisotropy, collisional quenching and resonance energy transfer are key techniques for protein detection.
  • Measuring the fluorescence decay allows the dynamics of structural changes in a protein to be observed directly. Moreover, excitation of the native fluorescence of proteins emanating from the amino acids tyrosine and tryptophan eliminates the possibility of perturbation of the local environment when using extrinsic fluorescent probes.
  • fluorescent probes for biological studies has been the use of naturally fluorescent proteins as fluorescent probes.
  • Naturally occurring dyes so-called fluorescent proteins (GFP, YFP, CFP, TOPAS, GFT, RFP), were discovered in the late 1990s (Clonetech, USA). These dyes are distinguished by their reduced influence on specimens. They are therefore particularly suitable for labeling cell regions in living preparations.
  • GFP green fluorescent protein
  • GFP green fluorescent protein
  • CFP cyan fluorescent protein
  • RFP red fluorescent protein
  • YFF yellow fluorescent protein
  • EYFP enhanced YFP
  • GFP GFP
  • variants of GFP, or other naturally occurring dyes may be coupled to non-natural amino acid polypeptides.
  • GFP can be used as a biosensor, reporting the results of levels of ions or pH by fluorescing in characteristic ways.
  • One molecule that can be used to sense the level of zinc ions is a blue fluorescent protein shown as PDB (Protein Data Bank) entry 1kys.
  • the protein fluoresces twice as brightly creating an easily detectable visible signal once zinc binds to the modified chromophore.
  • Construction of other peptide and protein biosensors comprising a non-natural amino acid may exhibit altered fluorescence properties in response to changes in their environment, oligomeric state, conformation upon ligand binding, structure, or direct ligand binding.
  • Appropriately labeled fluorescent biomolecules allow spatial and temporal detection of biochemical reactions inside living cells. See for example Giuliano, K.
  • probes are as following:
  • Labels Sensitivity and safety (compared to radioactive methods) of fluorescence has been increasingly used for specific labelling of nucleic acids, proteins and other biomolecules.
  • Fluorescein there are other fluorescent labels that cover the whole range from 400 to 820 nm.
  • some of the labels include, but are not limited to, Fluorescein and its derivatives, Carboxyfluoresceins, Rhodamines and their derivatives, Atto labels, Fluorescent red and Fluorescent orange: Cy3/Cy5TM alternatives, Lanthanide complexes with long lifetimes, Long wavelength labels - up to 800 nm, DY cyanine labels, Phycobili proteins.
  • Fluorescent molecules that are capable of absorbing radiation at one wavelength and emitting radiation at a longer wavelength include but are not limited to Alexa-532, Hydroxycoumarin, Aminocoumarin, Methoxycoumarin, Coumarin, Cascade Blue, Lucifer Yellow, P-Phycoerythrin, R-Phycoerythrin, (PE), PE-Cy5 conjugates, PE-Cy7 conjugates, Red 613, Fluorescein, BODIPY-FL, BODIPY TR, BODIPY TMR, Cy3, TRITC, X-Rhodamine, Lissamine Rhodamine B, PerCP, Texas Red, Cy5, Cy7, Allophycocyanin (APC), TruRed, APC-Cy7 conjugates, Oregon Green, Tetramethylrhodamine, Dansyl, Dansyl aziridine, Indo-1, Fura-2, FM 1-43, DilC18(3), Carboxy-SNARF-1, NBD, Indo-1, Fluo-3, DCFH, DHR,
  • Coumarin fluorescent dyes include, for example, amino methylcoumarin, 7-diethylamine-3-(4'-(1-maleimidyl)phenyl)-4-methylcoumarin (CPM) and N-(2-(1-maleimidyl)ethyl)7-diethylaminocoumarin-3-Carboxamide (MDCC).
  • Other useful molecules include those that display fluorescence resonance energy transfer (FRET).
  • FRET fluorescence resonance energy transfer
  • donor-acceptor pairs are known, and include fluorescein to rhodamine, coumarin to fluorescein or rhodamine, etc.
  • Still another class of useful label pairs includes fluorophore-quencher pairs in which the second group is a quencher, which decreases the fluorescence intensity of the fluorescent group.
  • Some known quenchers include acrylamide groups, heavy atoms such as iodide and bromate, nitroxide spin labels such as TEMPO, etc. Labels such as these may be
  • Fluorophores that are be conjugated to a non-natural amino acid polypeptide may fluoresce all of the time or only when the polypeptide is bound to a target.
  • Other types of fluorophores include
  • conjugates include but are not limited to, Isothiocyanate conjugates, streptavidin conjugates, and Biotin conjugates.
  • Antibody conjugates have been widely used to track biomolecules in living cells and whole organisms. They can be generated with specificity for virtually any epitope and are therefore, in principle, applicable to imaging a wide range of biomolecules.
  • Comjugates including but not limited to antibody conjugates may comprise a non-natural amino acid.
  • Enzyme substrates include but are not limited to fluorogenic and chromogenic substrates.
  • Micro - and Nanoparticles Various techniques allow the preparation of a wide variety of fluorescent microspheres ranging in size, matrix chemistry, type of fluorochrome, fluorescence intensity, and surface functional groups.
  • Fluorescent nanoparticles are promising tools for both optical data storage and other technical applications, for example, in biochemical, bioanalytical and medical areas.
  • Current medical and biological fluorescent imaging methods are mainly based on dye markers, which are limited in light emission per molecule, as well as photostability.
  • Nanoparticles overcome those problems offering strong and stable fluorescence. Fluorescent nanoparticles have been successfully used for various types of immunoassays. Fluorescent nanoparticles are based on different materials, such as, polyacrylonitrile, and polystyrene, etc.
  • Fluorescent Molecular Rotors are sensors of microenvironmental restriction that become fluorescent only if their rotation is constrained. The change of fluorescence intensity is caused by the restriction of intramolecular rotational relaxation about the donor-acceptor bond of the fluorophores. Examples of molecular constraint include but are not limited to increased dye (aggregation), binding to antibodies, or being trapped in the polymerization of actin.
  • IEF-Markers IEF (Isoelectric Focusing) is a powerful analytical tool for the separation of ampholytes, mainly proteins. In order to ensure the high performance of analysis, standards of pI (pI markers) are needed.
  • An advantage for IEF-Gel electrophoresis with Fluorescent IEF-Marker is the possibility to directly observe the formation of gradient. Fluorescent IEF-Marker can also be detected by UV-absorption at 280 nm (20°C).
  • FIG. 9 presents non-limiting examples of molecules that are site specifically attached to proteins through oxime formation between carbonyl of non-natural amino acid incorporated into a polypeptide and the hydroxylamine of the molecule.
  • the molecules shown are fluorophores, biotin, and chelators.
  • Bio-Orthogonal Chemical reporters Small molecules have better access to intracellular and extravascular compartments. Their use as imaging agents requires a means to selectively target the small probe to a desired biomolecule. Nucleophilic functionality occurs in most types of biopolymers, permitting facile derivatization with biotin, fluorophores and numerous other small-molecule reporters. Established bioconjugation protocols have made these operations trivial for purified biopolymers in vitro . It is an alternative strategy for tagging biomolecules that blends the simplicity of genetically encoded tags with the specificity of antibody labeling and the versatility of small-molecule probes. This approach involves the incorporation of unique chemical functionality—a bioorthogonal chemical reporter—into a target biomolecule using the cell's own biosynthetic machinery.
  • Bioorthogonal chemical reporters are non-native, non-perturbing chemical handles that can be modified in living systems through highly selective reactions with exogenously delivered probes. This two-step labeling process can be used to outfit a target biomolecule for detection or isolation, depending on the nature of the probe.
  • bio-orthogonal coupling reactions include but are not limited to, the Staudinger ligation of azides with triaryl phosphines, the ketone/ aldehyde-hydrazine reaction, and Huisgen's 1,3-dipolar azide-alkyne cycloaddition.
  • Replacement of the bulky fluorescent tag with a sterically inconspicuous azide group may furnish probes that are more able to distribute in an unbiased manner within a living cell, tissue, or organism.
  • the variable and often antagonistic effect of the fluorescent tag on probe binding affinity for specific proteins is also eliminated.
  • azide-alkyne cycloaddition chemistry can streamline probe synthesis by removing the need to generate and purify large quantities of structurally diverse fluorophore-tagged reagents. Coupling reactions utilizing non-natural amino acid polypeptides may provide probes that are alternatives to fluorescently tagged polypeptides. Huisgen's 1,3-dipolar azide-alkyne cycloaddition may be used to attach other molecules or provide other methods for polypeptide purification or detection.
  • Peptide libraries can be synthesized on solid supports and, by using coloring receptors, dyed solid supports can be selected one by one. If receptors cannot indicate any colors, their binding antibodies can be dyed. Because it is possible to separate solid supports by tweezers under microscopes or even magnifiers, the method can be not only be used on protein receptors, but also on screening binding ligands of synthesized artificial receptors and screening new metal binding ligands as well. This method is useful to search new lead compounds, because it enables the screening of a large amount of compounds.
  • FACS Fluorescence Activated Cell Sorter
  • This machine originally runs cells through a capillary tube and separates cells by detecting their fluorescent intensities. The same method may be used on solid supports instead of cells. Because it is designed for cells, small resins of cell size may be run, but normal sizes of solid supports (50 ⁇ 200 pmol) need specially modified machines. Partial or entire isolation of compounds may also be done. For partial isolation of compounds, time controlled photodecomposition or several functional groups to cleave in different conditions are used. In the meanwhile, one can scatter solid supports on soft agar and isolate some of compounds by photodecomposition. The isolated compounds then spread out around solid supports so that screening and solid support separation can be done at a time.
  • Methods for protein detection disclosed herein include immunoassays to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • Immunoassays combine the principles of chemistry and immunology enabling scientific tests, e.g. enzyme immunoassays and immunoblotting for a specific and sensitive detection of the analytes (non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof) of interest.
  • the basic principle of these assays is the specificity of the antibody-antigen reaction. Similar to the Western blot, a single protein can be identified by its antibody with immunoblotting.
  • the antigen may be non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof).
  • the antibodies or fragments thereof used in immunoassays may be non-natural amino acid polypeptides, and may be used in the detection of antigens that may or may not comprise a non-natural amino acid.
  • immunoassays are, by way of example only, RIAs (Radioimmunoassay) and enzyme immunoassays like ELISA (Enzyme-linked immunosorbent assay), EMIT (Enzyme Multiplied Immunoassay Technique), Microparticle Enzyme Immunoassay (MEIA), LIA (luminescent immunoassay), and FIA (fluorescent immunoassay).
  • ELISA Enzyme-linked immunosorbent assay
  • EMIT Enzyme Multiplied Immunoassay Technique
  • MEIA Microparticle Enzyme Immunoassay
  • LIA luminescent immunoassay
  • FIA fluorescent immunoassay
  • EMIT is a competitive binding immunoassay that avoids a separation step.
  • Methods for protein detection disclosed herein include ELISA to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • Enzyme linked immunosorbent assays are based on selective antibodies attached to solid supports combined with enzyme reactions to produce systems capable of detecting low levels of proteins. It is also known as enzyme immunoassay or EIA.
  • the antigen including but not limited to a protein, is detected by antibodies that have been made against it; that is, for which it is the antigen. Monoclonal antibodies are often used.
  • the test may require the antibodies to be fixed to a solid surface, such as the inner surface of a test tube; and a preparation of the same antibodies coupled to an enzyme.
  • the enzyme is one (e.g., ⁇ -galactosidase) that produces a colored product from a colorless substrate.
  • the test for example, is performed by filling the tube with the antigen solution (e.g., protein) to be assayed. Any antigen molecules present may bind to the immobilized antibody molecules.
  • the antibody-enzyme conjugate is added to the reaction mixture.
  • the antibody part of the conjugate binds to any antigen molecules that were bound previously, creating an antibody-antigen-antibody "sandwich". After washing away any unbound conjugate, the substrate solution is added.
  • the reaction is stopped (e.g., by adding 1 N NaOH) and the concentration of colored product formed by reaction of the substrate with molecules conjugated to the secondary antibody is measured in a spectrophotometer.
  • the intensity of color is proportional to the concentration of bound antigen.
  • ELISA can also be adapted to measure the concentration of antibodies, in which case, the wells are coated with the appropriate antigen.
  • the solution e.g., serum
  • an enzyme-conjugated anti-immunoglobulin is added, consisting of an antibody against the antibodies being tested for.
  • the substrate is added. The intensity of the color produced is proportional to the amount of enzyme-labeled antibodies bound (and thus to the concentration of the antibodies being assayed).
  • Radioimmunoassays to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • Radioactive isotopes can be used to study in vivo metabolism, distribution, and binding of small amount of compounds. Radioactive isotopes of 1 H, 12 C, 31 P, 32 S, 127 I are used such as 3 H, 14 C, 32 P, 35 S, 125 I. Radioactive isotopes have almost same chemical properties as unradioactive ones, so that they can be converted easily. Also because their radiation energy is relatively large, only a little amount is needed.
  • Receptor Fixation Method For a 96 well plate format, receptors are fixed in each well by using antibody or chemical methods, and radioactive labeled ligands are added to each well to induce binding. Unbound ligands are washed out and then the standard is determined by the quantitative analysis of the radioactivity of bound ligands or that of washed-out ligands. The addition of target compounds for screening induces competitive binding reactions with receptors. If target compounds show higher affinity to receptors than standard radioactive ligands, most of the radioactive ligands do not bind to receptors and are left in solution. Therefore, by analyzing the quantity of bound radioactive ligands (or washed-out ligands), the affinity of target compounds to receptors can be easily indicated.
  • a filter membrane method may be used when receptors cannot be fixed to 96 well plates or ligand binding must be performed in solution phase.
  • the reaction solution is filtered through nitrocellulose filter paper. Small molecules including ligands will go through the filter paper, and only protein receptors will be left on the paper. Only ligands that are strongly bound to receptors will stay on the filter paper, and the relative affinity of added compounds can be identified by quantitative analysis of the standard radioactive ligands.
  • This method can also be used to screen protein kinase inhibitors as well. In this case, ⁇ - 32 P-ATP can be used as a phosphoric acid group supplier, and by checking radioactive labeled protein substrate, enzymatic activity can be analyzed. Radioactive ATP that does not react will be filtered and removed.
  • radioimmunoassays can be performed by preparing a mixture of radioactive antigen and antibodies against that antigen. Iodine atoms can be introduced into tyrosine residues in a protein, the radioactive isotopes 125 I or 131 I are often used. Known amounts of unlabeled ("cold") antigen can be added to samples of the mixture. These compete for the binding sites of the antibodies. At increasing concentrations of unlabeled antigen, an increasing amount of radioactive antigen is displaced from the antibody molecules. The antibody-bound antigen is separated from the free antigen in the supernatant fluid, and the radioactivity of each is measured. From these data, a standard binding curve can be drawn. The samples to be assayed ("the unknowns”) are run in parallel. After determining the ratio of bound to free antigen in each unknown, the antigen concentrations can be read directly from the standard curve.
  • radioimmunoassays that can be used for detecting non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof are, by way of example only, precipitating the antigen-antibody complexes by adding a "second" antibody directed against the first.
  • a rabbit IgG is used to bind the antigen
  • the complex can be precipitated by adding an anti-rabbit-IgG antiserum (e.g., raised by immunizing a goat with rabbit IgG).
  • the antigen-specific antibodies can be coupled to the inner walls of a test tube.
  • the antigen-specific antibodies can be coupled to particles, like Sephadex. Centrifugation of the reaction mixture separates the bound counts (in the pellet) from the free counts in the supernatant fluid.
  • Methods for protein detection disclosed herein include fluorescence immunoassays to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof. Fluorescence based immunological methods are based upon the competitive binding of labeled ligands versus unlabeled ones on highly specific receptor sites. It is a very important tool for clinical and analytical biochemistry in the analysis of proteins.
  • This technique can be used for immunoassays based on changes in fluorescence lifetime with changing analyte concentration.
  • This technique works with dyes with a short lifetime like fluorescein isothiocyanate (FITC) (the donor) whose fluorescence is quenched by energy transfer to Eosin (the acceptor).
  • FITC fluorescein isothiocyanate
  • Eosin the acceptor
  • a number of molecular species have been used for causing energy transfer from a donor molecule to an acceptor molecule.
  • sandwich type immuno-complex formation can be used with this technique.
  • photoluminescent compounds may be used in the method disclosed herein and include the compounds listed above in fluorescence microscopy, as well as groups such as cyanines, oxazines, thiazines, porphyrins, phthalocyanines, fluorescent infrared-emitting polynuclear aromatic hydrocarbons, phycobiliproteins, squaraines and organo-metallic complexes, hydrocarbons and azo dyes.
  • Fluorescence based immunological methods can be, for example, heterogenous or homogenous.
  • Heterogenous Immunoassays comprise a physical separation of bound from free labeled analyte.
  • the analyte or antibody may be attached to a solid surface.
  • the technique can be competitive (for a higher selectivity) or noncompetitive (for a higher sensitivity).
  • Detection can be direct (only one type of antibody used) or indirect (a second type of antibody is used).
  • Homogenous Immunoassays comprise no physical separation. Double-Antibody Fluorophore-labeled antigen participates in an equilibrium reaction with antibodies directed against both the antigen and the fluorophore. Labeled and unlabeled antigens compete for a limited number of anti-antigen antibodies.
  • Simple Fluorescence Labelling method It can be used for receptor-ligand binding, enzymatic activity by using pertinent fluorescence, and as a fluorescent indicator of various in vivo physiological changes such as pH, ion concentration, and electric pressure. Self-fluorescence of amino acids such as tyrosine and tryptophan result in background radiation, and to overcome such weak points fluorescent compounds of absorption UV length longer than 520 nm such as cyanine are often used.
  • FRET Fluorescence Resonance Energy Transfer - FRET may be used to measure the interaction of two proteins in vivo and can measure nanometer scale distances and distance (conformation) changes. Therefore, it has been used to measure simple protein-protein interactions and changes in protein folding, conformation, and stability (see Philipps, B.; Hennecke, J.; Glockshuber R. Mol Biol. 2003, 327, 239-249 ; Riven, I.; Kalmanzon, E.; Segev, L.; Reuveny E. Neuron. 2003, 38, 225-235 ).
  • Two different fluorescent molecules (fluorophores) are conjugated to the two proteins of interest. Non-natural amino acid polypeptides conjugated to fluorophores may be used in FRET.
  • Distance R0 of 50% FRET effect depends on the overlap of the emission range of donors, absorption range of the acceptors, and the acceptor's quantum yields and solvent. If two fluorescent molecules are at a shorter distance from each other than R0, when the donor's absorption light is emitted, theoretically the acceptor's fluorescence will be stronger. If the distance becomes longer than R0, when the same light is emitted, the donor's fluorescence will be detected as stronger. Therefore, enzymatic activity can be measured easily if fluorescent molecules are linked to the ends of small peptides, which can be used as kinases such as protease.
  • BRET Bioluminescene resonance energy transfer
  • TRF Time Resolved Fluorescence - To reduce fluorescent background, Time Resolved Fluorescence was developed. The lifetime of excited states of common fluorescent molecules is usually only a few microseconds, but Lanthanide series elements have milliseconds of life time. TRF is a method that selectively measures the fluorescence of the Lanthanide series after the emission of other fluorescent molecules has finished. TRF can be also with FRET, and Lanthanide series become donors or acceptors.
  • Various assay formats may be used for the detection of the non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides, and fragment thereof, disclosed herein, including "sandwich" immunoassays and probe assays.
  • a polyclonal or monoclonal antibody or fragment thereof, or a combination of these antibodies which has been coated on a solid phase, is contacted with a test sample, to form a first mixture. This first mixture is incubated for a time and under conditions sufficient to form antigen/antibody complexes.
  • an indicator reagent comprising a monoclonal or a polyclonal antibody or a fragment thereof, or a combination of these antibodies, to which a signal generating compound has been attached, is contacted with the antigen/antibody complexes to form a second mixture.
  • This second mixture then is incubated for a time and under conditions sufficient to form antibody/antigen/antibody complexes.
  • the presence of antigen in the test sample and captured on the solid phase, if any, is determined by detecting the measurable signal generated by the signal generating compound.
  • the amount of antigen present in the test sample is proportional to the signal generated.
  • a mixture is formed by contacting: (1) a polyclonal antibody, monoclonal antibody, or fragment thereof, which specifically binds to antigen, or a combination of such antibodies bound to a solid support; (2) the test sample; and (3) an indicator reagent comprising a monoclonal antibody, polyclonal antibody, or fragment thereof, which specifically binds to a different epitope (or a combination of these antibodies) to which a signal generating compound is attached.
  • This mixture is incubated for a time and under conditions sufficient to form antibody/antigen/antibody complexes.
  • the presence, if any, of antigen present in the test sample and captured on the solid phase is determined by detecting the measurable signal generated by the signal generating compound.
  • the amount of antigen present in the test sample is proportional to the signal generated.
  • one or a combination of at least two monoclonal antibodies of the invention can be employed as a competitive probe for the detection of antibodies to antigen.
  • unnatural amino acid polypeptides disclosed herein, either alone or in combination are coated on a solid phase.
  • a test sample suspected of containing antibody to antigen then is incubated with an indicator reagent comprising a signal generating compound and at least one monoclonal antibody for a time and under conditions sufficient to form antigen/antibody complexes of either the test sample and indicator reagent bound to the solid phase or the indicator reagent bound to the solid phase.
  • the reduction in binding of the monoclonal antibody to the solid phase can be quantitatively measured.
  • the monoclonal or polyclonal antibodies can be employed in the detection of antigens in tissue sections, as well as in cells, by immunohistochemical analysis.
  • tissue sections can be cut from either frozen or chemically fixed samples of tissue.
  • the cells can be isolated from blood, urine, breast aspirates, or other bodily fluids.
  • the cells may be obtained by biopsy, either surgical or by needle.
  • the cells can be isolated by centrifugation or magnetic attraction after labeling with magnetic particles or ferrofluids so as to enrich a particular fraction of cells for staining with the antibodies.
  • Combinations of the monoclonal antibodies (and fragments thereof) also may be used together as components in a mixture or "cocktail" along with antibodies which specifically bind to other regions of unnatural amino acid polypeptides disclosed herein, each antibody having different binding specificities.
  • the polyclonal antibodies used in the assays can be used either alone or as a cocktail of polyclonal antibodies. Since the cocktails used in the assay formats are comprised of either monoclonal antibodies or polyclonal antibodies having different binding specificity to unnatural amino acid polypeptides disclosed herein, they are useful for the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining the predisposition to, various diseases and conditions.
  • unnatural amino acid amino acids disclosed herein may be detected in assays by use of a recombinant antigen as well as by use of a synthetic polypeptide or purified polypeptide, which polypeptide comprises an amino acid sequence of unnatural amino acid polypeptides disclosed herein. It also contemplated that different synthetic, recombinant or purified polypeptides, identifying different epitopes of unnatural amino acid polypeptides disclosed herein, can be used in combination in an assay for the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, etc.
  • polypeptides can be coated onto one solid phase; or each separate polypeptide may be coated onto separate solid phases, such as microparticles, and then combined to form a mixture of polypeptides which can be later used in assays.
  • Polypeptides coated on solid phases or labeled with detectable labels are then allowed to compete with those present in a sample for a limited amount of antibody.
  • a reduction in binding of the synthetic, recombinant, or purified peptides to the antibody (or antibodies) is an indication of the presence of unnatural amino acid polypeptides disclosed herein. Variations of assay formats are known to those of ordinary skill in the art.
  • Methods for protein detection disclosed herein include SPM to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • scanning probe microscopy in the capture phase, for example, at least one of the monoclonal antibodies is adhered to a solid phase and a scanning probe microscope is utilized to detect antigen/antibody complexes which may be present on the surface of the solid phase.
  • the use of scanning tunneling microscopy eliminates the need for labels which normally must be utilized in many immunoassay systems to detect antigen/antibody complexes.
  • one member of a specific binding partner is attached to a surface suitable for scanning.
  • the attachment of the analyte specific substance may be by adsorption to a test piece which comprises a solid phase of a plastic or metal surface.
  • Covalent attachment of a specific binding partner (analyte specific substance) to a test piece which test piece comprises a solid phase of derivatized plastic, metal, silicon, or glass may be utilized. Covalent attachment methods are known to those skilled in the art and include a variety of means to irreversibly link specific binding partners to the test piece.
  • the surface must be activated prior to attaching the specific binding partner.
  • polyelectrolyte interactions may be used to immobilize a specific binding partner on a surface of a test piece by using techniques and chemistries. The preferred method of attachment is by covalent means.
  • the surface may be further treated with materials such as serum, proteins, or other blocking agents to minimize non-specific binding.
  • the surface also may be scanned either at the site of manufacture or point of use to verify its suitability for assay purposes. The scanning process is not anticipated to alter the specific binding properties of the test piece.
  • Methods for protein detection disclosed herein include NMR to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • NMR spectroscopy is capable of determing the structures of biological macromolecules like proteins and nucleic acids at atomic resolution. In addition, it is possible to study time dependent phenomena with NMR, such as intramolecular dynamics in macromolecules, reaction kinetics, molecular recognition or protein folding. Methods for protein detection disclosed herein, include NMR to detect non-natural amino acid polypeptides and modified non-natural amino acid polypeptides and fragments thereof.
  • Methods for protein detection disclosed herein include X-ray crystallography to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • X-ray crystallography is a technique in crystallography in which the pattern produced by the diffraction of X-rays through the closely spaced lattice of atoms in a crystal is recorded and then analyzed to reveal the nature of that lattice. This generally leads to an understanding of the material and molecular structure of a substance. The spacings in the crystal lattice can be determined using Bragg's law. The electrons that surround the atoms, rather than the atomic nuclei themselves, are the entities which physically interact with the incoming X-ray photons. This technique is widely used in chemistry and biochemistry to determine the structures of an immense variety of molecules, including inorganic compounds, DNA and proteins. X-ray diffraction is commonly carried out using single crystals of a material, but if these are not available, microcrystalline powdered samples may also be used, although this requires different equipment and is much less straightforward.
  • the molecule For X-ray crystallography, the molecule must be crystallized. One photon diffracted by one electron cannot be reliably detected, however, because of the regular crystalline structure; the photons are diffracted by corresponding electrons in many symmetrically arranged molecules. Because waves of the same frequency whose peaks match reinforce each other, the signal becomes detectable.
  • crystals of the molecule of interest are grown using some method of crystallization. The crystals are harvested and often frozen with liquid nitrogen. Freezing crystals both reduces radiation damage incurred during data collection and decreases thermal motion within the crystal. Crystals are placed on a diffractometer, a machine that emits a beam of X-rays.
  • the X-rays diffract off the electrons in the crystal, and the pattern of diffraction is recorded on film and scanned into a computer. These diffraction images are combined and eventually used to construct a map of the electron density of the molecule that was crystallized, atoms are then fitted to the electron density map and various parameters such as position are refined to best fit the observed diffraction data.
  • Methods for protein detection disclosed herein include fluorescence spectroscopy to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • Fluorescence Polarization polarized light is used for excitation and binding of fluorochrome-labeled antigens to specific antibodies affects polarization extent.
  • Line Narrowing Spectroscopy involves low-temperature solid-state spectroscopy that derives its selectivity from the narrow-line emission spectra it provides.
  • Time-dependent Fluorescence Spectroscopy comprises time-resolved measurements containing more information than steady-state measurements, since the steady-state values represent the time average of time-resolved determinations. It is a single photon timing technique in which the time between an excitation light pulse and the first photon emitted by the sample is measured.
  • Frequency-Domain Fluorescence Spectroscopy is an alternative to the time-resolved methods.
  • the time decay of fluorescence is typically measured using a light source with an intensity modulated sinusoidally at a given frequency, by determining the phase delay and the relative modulation of the fluorescence signal with respect to the exciting light.
  • MALDI TOF-MS Matrix Assisted Laser Desorption ionization time-of-flight mass spectrometry
  • Methods for protein detection disclosed herein include MALDI TOF-MS to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • kinetic energy
  • m the mass of the ion
  • v velocity of the ion
  • z number of charges
  • e the charge on an electron in coulombs
  • E electric field gradient
  • s the distance of the ion source region
  • Reflectron TOF-MS- Improved mass resolution in MALDI TOF-MS has been obtained by the utilization of a single-stage or a dual-stage reflectron (RETOF-MS).
  • the reflectron located at the end of the flight tube, is used to compensate for the difference in flight times of the same m/z ions of slightly different kinetic energies by means of an ion reflector. This results in focusing the ion packets in space and time at the detector.
  • the isotopic multiplet is well resolved producing a full width half maximum (FWHM) mass resolution of about 3400.
  • Mass resolutions up to 6000 (FWHM) have been obtained for peptides up to about 3000 Da with RETOF-MS. Enhancing the mass resolution can also increase the mass accuracy when determining the ion's mass.
  • both linear and reflectron MALDI-TOF-MS have been utilized primarily for molecular weight determinations of molecular ions and enzymatic digests leading to structural information of proteins. These digests are typically mass analyzed with or without purification prior to molecular weight determinations. Varieties of methodologies have been developed to obtain primary sequence information for proteins and peptides utilizing MALDI TOF-MS. Two different approaches can be taken. The first method is known as protein ladder sequencing and is employed to produce structurally informative fragments of the analyte prior to insertion into the TOF mass spectrometer and subsequent analysis. The second approach utilizes the phenomenon of metastable ion decay that occurs inside the TOF mass spectrometer to produce sequence information.
  • Ladder Sequencing with TOF-MS -Proteins or peptides can be sequenced using MALDI -TOF-MS with a ladder sequencing technique which consists of either a time-dependent or concentration-dependent chemical degradation from either the N- or C-terminus of the protein or peptide into fragments, each of which differs by one amino acid residue.
  • the mixture is mass analyzed in a single MALDI -TOF-MS experiment with mass differences between adjacent mass spectral peaks corresponding to a specific amino acid residue. This type of analysis can be thought of as simply determining the masses of a series of peptides/proteins that are present in a single MALDI sample. The order of occurrence in the mass spectrum defines the sequence of amino acids in the original protein or peptide.
  • MALDI TOF-MS has developed into a valuable tool in the biosciences for obtaining both accurate mass determinations and primary sequence information.
  • Methods for protein detection disclosed herein include MALDI TOF-MS to detect non-natural amino acid polypeptides and modified non-natural amino acid polypeptides and fragments thereof.
  • sequence information obtained from the mass spectra whose sequence was known a priori by no means implies a straightforward scheme to deduce an unknown peptide or protein sequence from its metastable ion decay mass spectrum.
  • These MALDI techniques are envisioned to be most useful in conjunction with conventional biochemical techniques such as protein digests. They may be applicable to identifying blocked amino termini, post-translational modifications and mutation sites in known proteins in this way. Also, with a total unknown, a significant amount of preliminary structure determination should be possible on very small (less than 10 pmol) amounts of analyte. For ladder sequencing and in-source fragmentation studies, it is important to minimize potential peptide impurities.
  • SELDI-TOF surface-enhanced laser desorption ionization - time of flight
  • This technique utilizes stainless steel or aluminum-based supports, or chips, engineered with chemical (hydrophilic, hydrophobic, pre-activated, normal-phase, immobilized metal affinity, and cationic or anionic) or biological (antibody, antigen binding fragments (including but not limited to, scFv), DNA, enzyme, or receptor) bait surfaces of 1-2mm in diameter.
  • chemical hydrophilic, hydrophobic, pre-activated, normal-phase, immobilized metal affinity, and cationic or anionic
  • biological antibody, antigen binding fragments (including but not limited to, scFv), DNA, enzyme, or receptor) bait surfaces of 1-2mm in diameter.
  • Solubilized tissue or body fluids in volumes as small as 0.1 1 g are directly applied to these surfaces, where proteins with affinities to the bait surface will bind. Following a series of washes to remove non-specifically or weakly bound proteins, the bound proteins are laser desorbed and ionized for MS analysis.
  • Masses of proteins ranging from small peptides of less than 1000 Da up to proteins of greater than 300 kDa are calculated based on time-of-flight. As mixtures of proteins will be analyzed within different samples, a unique sample fingerprint or signature will result for each sample tested. Consequently, patterns of masses rather than actual protein identifications are produced by SELDI analysis. These mass spectral patterns are used to differentiate patient samples from one another, such as diseased from normal. While protein fingerprints can be analyzed for differential biomarker expression, this technology is currently unable to specifically identify proteins within a sample using MS. However, this situation is rapidly evolving as prototypes are being tested which couple the SELDI-TOF technology with tandem mass spectrometers. Coupling of these types of instruments will enable amino acid sequencing and subsequent protein identification.
  • Methods for protein detection disclosed herein include UV-Vis to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • UV/VIS Optical absorption spectroscopy
  • Resonance Raman Spectroscopy is a tool which can be used to study molecular structure and dynamics.
  • Resonance Raman scattering requires excitation within an electronic absorption band and results in a large increase of scattering. Few molecules have visible absorption bands; however everything absorbs in the deep UV. By using UV light it is possible to study a wide variety of colorless chromophores, and have the additional benefit of avoiding interference from fluorescence.
  • electrons of different functional groups with different excitation wavelengths can be selectively excited. This approach helps to investigate specific parts of macromolecules by using different excitation wavelengths.
  • Liquid chromatography has been a powerful tool for isolating proteins, peptides, and other molecules from complex mixtures.
  • Methods for protein detection disclosed herein include LC to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • Liquid chromatography can be affinity chromatography, gel filtration chromatography, anion exchange chromatography, cation exchange chromatography, diaode array -LC and high performance liquid chromatography (HPLC).
  • Gel filtration chromatography seprarates proteins, peptides, and oligonucleotides on the basis of size. Molecules move through a bed of porous beads, diffusing into the beads to greater or lesser degrees. Smaller molecules diffuse further into the pores of the beads and therefore move through the bed more slowly, while larger molecules enter less or not at all and thus move through the bed more quickly. Both molecular weight and three dimensional shape contribute to the degree of retention. Gel Filtration Chromatography may be used for analysis of molecular size, for separations of components in a mixture, or for salt removal or buffer exchange from a preparation of marcromolecules.
  • Affinity chromatography is the process of bioselective adsorption and subsequent recovery of a compound from an immobilized ligand. This process allows for the highly specific and efficient purification of many diverse proteins and other compounds.
  • the process requires the utilization of an appropriately selective ligand which will bind the desired compound generally with a dissociation constant in the range of 10 4 to 10 -8 , while permitting recovery under mild conditions.
  • the ligand is generally immobilized on a beaded and porous matrix which may be in the form of a column packing or batchwise adsorption medium.
  • Ion exchange chromatography separates molecules based on differences between the overall charge of the proteins. It is usually used for protein purification but may be used for purification of oligonucleotides, peptides, or other charged molecules.
  • the protein of interest must have a charge opposite that of the functional group attached to the resin in order to bind.
  • immunoglobulins which generally have an overall positive charge, will bind well to cation exchangers, which contain negatively charged functional groups. Because this interaction is ionic, binding must take place under low ionic conditions. Elution is achieved by increasing the ionic strength to break up the ionic interaction, or by changing the pH of the protein.
  • HPLC can be used in the separation, purification and detection of non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof disclosed herein.
  • Peptides Use of reversed-phased chromatography (RPC) has become a common and important step in synthetic peptide production. RPC has also been used to purify natural sequences. Although analytical columns are used to carry out the process, the procedure can be preparative in nature due to the limited amount of "active" proteins in tissue.
  • Crude tissue extracts may be loaded directly onto the RPC system and mobilized by gradient elution. Rechromatography under the identical conditions is an option if further purification is warranted or necessary.
  • RPC can also be utilized in the process of protein structure determination. The normal procedure of this process is 1) fragmentation by proteolysis or chemical cleavage; 2) purification; and 3) sequencing.
  • a common mobile phase for RPC of peptides is a gradient of 0.1% trifluoroacetic acid (TFA) in water to 0.1% TFA in an organic solvent, such as acetonitrile, since the organic solvent 1) solubilizes the peptide, 2) allows detection at approximately 230-240 nm, and 3) can evaporate away from the sample.
  • TFA trifluoroacetic acid
  • Biologically Active Proteins The use of size-exclusion chromatography (SEC) and ion-exchange chromatography (IEC) is well-suited for use with biologically active proteins, such as enzymes, hormones, and antibodies, since each protein has its own unique structure and the techniques may be performed in physiological conditions. Full recovery of activity after exposure to the chromatography may be achieved, and currently, availability of SEC columns is diverse enough to allow fractionation from 10 to 1000 kilodaltons. Extremely basic or hydrophobic proteins may not exhibit true SEC character since the columns tend to have slight hydrophobicity and anionic character.
  • Membrane Proteins are either peripheral (situated on the outer surface) or integral (partially span, entirely span, or lie completely within the membrane). The lipophilicity of the bilayer conveys the lipophilic character (i.e., hydrophobic amino acids) of the proteins within the membrane.
  • RPC would be a logical choice in analysis and purification of these proteins, but IEC is also employed.
  • Another procedure used in the separation of membrane proteins is the use of nonionic detergents, such as Triton X-100, or protein solubilization by organic solvents with IEC.
  • HPLC may be coupled with MS.
  • Diode array detector-liquid chromatography provides complete, multiple spectra for each HPLC peak, which, by comparison, can provide indication of peak purity. These data can also assign presence of Tyr, Trp, Phe, and possibly others (His, Met, Cys) and can quantitate these amino acids by 2nd derivative or multi-component analysis. By a post-column derivatization, DAD-LC can also identify and quantitate Cys, His and Arg in individual peptides. Thus, it is possible to analyze for 6 of the 20 amino acids of each separated peptide in a single LC run, and information can be obtained about presence or absence of these amino acids in a given peptide in a single step. This is assisted by knowing the number of residues in each peptide. Also, by correction at 205 nm absorbance for side-chain chromophores, this technique can give much better estimation of relative amounts of each peptide.
  • Electrophoresis to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof Electrophoresis can be gel electrophoresis or capillary electrophoresis.
  • Gel electrophoresis is a technique that can be used for the separation of proteins. Separation of large (macro) molecules depends upon two forces: charge and mass. When a biological sample, such as proteins, is mixed in a buffer solution and applied to a gel, these two forces act together. The electrical current from one electrode repels the molecules while the other electrode simultaneously attracts the molecules. The frictional force of the gel material acts as a "molecular sieve,” separating the molecules by size. During electrophoresis, macromolecules are forced to move through the pores when the electrical current is applied.
  • Electrofocusing or isoelectric focusing, is a technique for separating different molecules by their electric charge differences (if they have any charge). It is most commonly used on proteins. It is a type of zone electrophoresis that takes advantage of the fact that a molecule's charge changes as the pH of its surroundings changes. Molecules are distributed over a medium that has a pH gradient (usually created by aliphatic ampholytes). An electric current is passed through the medium, creating a "positive” and “negative” end. Negatively charged particles migrate through the pH gradient toward the "positive” end while positively charged particles move toward the "negative” end. As a particle moves into a pH that neutralizes its charge, it will stop following the current. Particles of the same initial charge will deposit (or focus) around the same place on the pH gradient.
  • Capillary electrophoresis is a collection of a range of separation techniques which involve the application of high voltages across buffer filled capillaries to achieve separations. The variations include separation based on size and charge differences between analytes (termed Capillary Zone Electrophoresis, CZE, or Free Solution CE, FSCE), separation of neutral compounds using surfactant micelles (Micellar electrokinetic capillary chromatography, MECC or sometimes referred to as MEKC) sieving of solutes through a gel network (Capillary Gel Electrophoresis, GCE), separation of cations (or anions) based on electrophoretic mobility (Capillary Isotachophoresis, CITP), and separation of zwitterionic solutes within a pH gradient (Capillary Isoelectric Focusing, CIEF).
  • CZE Capillary Zone Electrophoresis
  • FSCE Free Solution CE
  • MECC Licellar electrokinetic capillary chromatography
  • MEKC Microcellar electrokinetic capillar
  • Capillary electrochromatography is an associated electrokinetic separation technique which involves applying voltages across capillaries filled with silica gel stationary phases. Separation selectivity in CEC is a combination of both electrophoretic and chromatographic processes. Many of the CE separation techniques rely on the presence of an electrically induced flow of solution (electroosmotic flow, EOF) within the capillary to pump solutes towards the detector. GCE and CIEF are of importance for the separation of biomolecules such as proteins. Generally CE is performed using aqueous based electrolytes however there is a growing use of nonaqueous solvents in CE.
  • Operation of a CE system involves application of a high voltage (typically 10-30kV) across a narrow bore (25-100mm) capillary.
  • the capillary is filled with electrolyte solution which conducts current through the inside of the capillary.
  • the ends of the capillary are dipped into reservoirs filled with the electrolyte.
  • Electrodes made of an inert material such as platinum are also inserted into the electrolyte reservoirs to complete the electrical circuit.
  • a small volume of sample is injected into one end of the capillary.
  • the capillary passes through a detector, usually a UV absorbance detector, at the opposite end of the capillary.
  • Application of a voltage causes movement of sample ions towards their appropriate electrode usually passing through the detector.
  • electropherogram The plot of detector response with time is generated which is termed an electropherogram.
  • a flow of electrolyte known as electroendosmotic flow, EOF, results in a flow of the solution along the capillary usually towards the detector. This flow can significantly reduce analysis times or force an ion to overcome its migration tendency towards the electrode it is being attracted to by the sign of its charge.
  • Methods for protein detection disclosed herein include arrays to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof.
  • Microarrays involve performing parallel analysis of multiple samples against known protein targets.
  • the development of various microarray platforms has remarkably enabled and accelerated the determination of protein abundance, localization, and interactions in a cell or tissue.
  • Microarrays provide a platform that allows identification of protein interaction or function against a characterized set of proteins, antibodies, or peptides.
  • Protein-based chips array proteins on a small surface and can directly measure the levels of proteins in tissues using fluorescence-based imaging. Proteins can be arrayed on either flat solid phases or in capillary systems (micro fluidic arrays), and several different proteins can be applied to these arrays. The most popular ones currently rely on antibody-antigen interactions, which can also detect antigen-protein interactions. The potential of antibody arrays is currently limited by the availability of antibodies that have both high specificity (to eliminate cross reactions with non-specific proteins within the sample) and high affinity for the target of interest (to allow detection of small quantities within a sample). Another challenge of protein array technology is the ability to preserve proteins in their biologically active shape and form.
  • oligonucleotides In addition to the use of antibodies as array probes, single-stranded oligonucleotides, whose specificity is optimized by in vitro elution (aptamers), offer a viable alternative. Aptamers allow their covalent attachment to cognate proteins by photo-crosslinking, thus reducing background. Nonspecific protein stains are then used to detect bound proteins.
  • Arrays include, but not limited to, bead arrays, bead based arrays, bioarrays, bioelectronic arrays, cDNA arrays, cell arrays, DNA arrays, gene arrays, gene expression arrays, frozen cell arrays, genome arrays, high density oligonucleotide arrays, hybridization arrays, microcantilever arrays, microelectronic arrays, multiplex DNA hybridization arrays, nanoarrays, oligonucleotide arrays, oligosaccharide arrays, planar arrays, protein arrays, solution arrays, spotted arrays, tissue arrays, exon arrays, filter arrays, macroarrays, small molecule microarrays, suspension arrays, theme arrays, tiling arrays, and transcript arrays.
  • Methods for protein detection disclosed herein include sensors to detect non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof. Sensors can be used for both in vivo and in vitro detection. Sensors may be used to detect events such as binding of a non-natural amino acid polypeptide to its target, conformational changes in a non-natural amino acid polypeptide, and or measure other interactions, modifications, or changes to a non-natural amino acid polypeptide or its environment.
  • Sensors can be chemical sensors, optical sensors, and biosensors.
  • Chemical sensors are miniaturized analytical devices which deliver real-time and online information on the presence of specific compounds or ions in complex samples.
  • Optical sensors are based on measurement of either intrinsic optical properties of analytes, or of optical properties of indicator dyes or labeled biomolecules attached to solid supports.
  • Biosensors can be affinity biosensor based on capabilities of enzymes to convert "substrates" into products; or catalytic biosensors.
  • the binding of a non-natural amino acid polypeptide to its target including but not limited to, an antibody, antibody fragment, or antigen-binding polypeptide or fragment thereof, may be measured.
  • the non-natural amino acid polypeptide is conjugated to a molecule such as a nanotransmitter. While bound to its target in-vivo, the nanotransmitter emits a signal that is read ex vivo by a medical imaging instrument.
  • Protein separation aids to separate a complex mixture so that individual proteins are more easily processed with other techniques.
  • Protein identification methods include but is not limited to low-throughput sequencing through Edman degradation, mass spectrometry techniques, peptide mass fingerprinting, de novo sequencing, antibody-based assays and protein quantification assays such as fluorescent dye gel staining, tagging or chemical modification methods ( i.e. isotope-coded affinity tags - ICATS, combined fractional diagonal chromatography - COFRADIC).
  • the purified protein may also be used for determination of three-dimensional crystal structure, which can be used for modeling intermolecular interactions. Common methods for determining three-dimensional crystal structure include x-ray crystallography and NMR spectroscopy. Detailed below are a few of the methods for identifying proteins.
  • N-terminal sequencing aids in the identification of unknown proteins; confirm recombinant protein identity and fidelity (reading frame, translation start point, etc .); aid the interpretation of NMR and crystallographic data; demonstrate degrees of identity between proteins; or provide data for the design of synthetic peptides for antibody generation, etc.
  • N-terminal sequencing utilizes the well-established Edman degradative chemistry, sequentially removing amino acid residues from the N-terminus of the protein and identifying them by reverse-phase HPLC. Sensitivity is at the level of 100s femtomoles and long sequence reads (20-40 residues) can often be obtained from a few 10s picomoles of starting material.
  • Pure proteins (>90%) generate easily interpreted data, but insufficiently purified protein mixtures may also provide useful data, subject to rigorous data interpretation.
  • N-terminally modified (especially acetylated) proteins cannot be sequenced directly, as the absence of a free primary amino-group prevents the Edman chemistry.
  • limited proteolysis of the blocked protein e.g. using cyanogen bromide
  • C-terminal sequencing is recognized as an important post-translational modification, sometimes critically affecting the structure and activity of a protein.
  • Various disease situations have been associated with impaired protein processing and C-terminal sequencing provides an additional tool for the investigation of protein structure and processing mechanisms.
  • proteomics proteins can be identified primarily by computer search algorithms that assign sequences to a set of empirically acquired mass/intensity data which are generated from conducting electrospray ionization (ESI), matrix-assisted laser desorption/ionization (MALDI), time-of-flight (TOF) instruments, or a three-dimensional quadrupole ion traps on the protein of interest.
  • ESI electrospray ionization
  • MALDI matrix-assisted laser desorption/ionization
  • TOF time-of-flight
  • Additional detection methods involve bipyridines, metal coordination, nanotechnology (gold), biotin-streptavidin/avidin, UV/Vis, 2 step systems that involve a binding event and a coupling event due to proximity of a non-natural amino acid to a target resulting in exmission from a fluorophore, small molecule based fluorescent/fluorogenic molecules bound to a non-natural amino acid present in a polypeptide, lipocalins (beta barrel), fatty acid binding proteins, and dark to light or light to dark fluorophores.
  • Molecular Imaging is a multidisciplinary field involving the efforts from molecular and cell biology to identify the molecular imaging target, radiochemistry and bioconjugation chemistry to develop suitable imaging probes, pharmacology to optimize the probes for optimal targeting efficacy and favorable in vivo kinetics, and image-capture techniques to non-invasively monitor the fate of molecular imaging probes in vivo .
  • molecular imaging also plays roles in treatment efficacy assessment, drug discovery, and understanding of molecular mechanisms in living systems.
  • Molecular imaging probes can be used for visualization and quantification of molecular targets.
  • microMRI and microCT anatomical
  • microPET micropositron emission tomography
  • microSPECT micropositron emission tomography
  • NIR fluorescence imaging NIR fluorescence imaging
  • Bio-imaging methods can be used to detect spatial organization (i.e., distribution) and to quantify cellular and tissue natural constituents, structures, organelles and administered components such as tagging probes (e.g., fluorescent probes) and drugs using light transmission, reflection, scattering and fluorescence emission strategies, with high spatial and spectral resolutions.
  • tagging probes e.g., fluorescent probes
  • In-vivo competition assays of unlabeled compounds with labeled probes for agents with known pharmacological characteristics and efficacy can be used in the drug evaluation process.
  • Noninvasive characterization of drug targeting, receptor occupancy, concentrations required for effective receptor or enzyme inhibition, etc. can speed up the evaluation of lead compounds.
  • imaging analyses can quantitatively and repetitively monitor target accessibility, duration of retention at the target site and its correlation with drug efficacy, and clearance from irrelevant tissues.
  • imaging assays can facilitate evaluation of non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof, for both their pharmacological properties and their therapeutic effectiveness in patients.
  • physicians can perform multiple functional-imaging assays simultaneously with anatomic analyses.
  • Information derived from structural studies and from noninvasive, repetitive monitoring of drug distribution and concentration can then be correlated with biological effects on signal transduction pathways, target enzyme activities, antigen levels, receptor activation, cell proliferation, proteasome activity, etc.
  • These noninvasive assays can permit real-time monitoring and modification of targeted interventions and therapeutic strategies.
  • Molecular-imaging technologies can be used to study mouse models in pre-clinical studies.
  • many drugs for cancer and other disorders exert their therapeutic effects by inducing apoptosis.
  • the ability to repetitively image apoptotic responses in living animals can facilitate preclinical evaluation of these drugs.
  • identification of founder mice that can express the transgene in the proper spatial and temporal pattern by noninvasive imaging can permit the identification of founders without breeding.
  • Molecular imaging can provide the location, magnitude, and duration of expression of the therapeutic gene for the optimization of gene-therapy protocols. Optical imaging can be coupled with targeted gene transfer. Molecular imaging of reporter genes can also be used to monitor the biodistribution and efficacy of cell-based therapies.
  • Imaging probes can be molecules labeled with radioisotopes or light- or nearinfrared (NIR)-emitting molecules. The concentration and/or spectral properties of molecular imaging probes are altered by the specific biological process under investigation.
  • Two types of probes that can be used in functional imaging studies are, by way of example only, direct binding probes and indirect probes.
  • Direct binding probes and indirect probes may be non-natural amino acid polypeptides. Examples of direct binding probes include but are not limited to antibodies, antibody fragments, antigen-binding polypeptides and fragments thereof and receptor ligands. Direct probes can be used to detect concentrations of their targets, since their binding is stochiometric.
  • direct probes are useful in investigating targets that are overexpressed in pathological conditions, for example, before and after therapy.
  • Indirect probes are used to monitor activities of their macromolecular targets, including catalytic activities. Examples of such probes are described by Herschman in Science 2003 302:605-608 .
  • Probes can be developed to monitor endogenous targeted molecules and biological processes. Such probes may be (modified) non-natural amino acid polypeptides. Key mediators and/or indicators of endogenous processes may be investigated using imaging probes. Substrates for enzymes such as kinases or proteases may be labeled via radionuclides or fluorescent molecules such that events such as phosphorylation or protease cleavage are detected by molecular-imaging assays. Such fluorescent probes that emit NIR fluorescent light after protease cleavage may be referred to as "activatable" optical imaging probes.
  • Direct and indirect probes may be discovered by high-throughput screening of chemical libraries. Direct probes may also be discovered by screening large recombinant antibody and phage libraries. Such libraries may be composed of (modified) non-natural amino acid polypeptides.
  • Quantum dots Methods for imaging and diagnostics utilizing non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof disclosed herein include fluorescent semiconductor nanocrystals (also known as quantum dots or qdots). Qdots can be used for the study of intracellular processes at the single-molecule level, high-resolution cellular imaging, long-term in vivo observation of cell trafficking, tumor targeting, and diagnostics.
  • fluorescent semiconductor nanocrystals also known as quantum dots or qdots
  • Colloidal semiconductor quantum dots are single crystals a few nanometers in diameter whose size and shape can be precisely controlled by the duration, temperature, and ligand molecules used in the synthesis. This process may yield qdots that have composition-and size-dependent absorption and emission. Absorption of a photon with energy above the semiconductor band gap energy may result in the creation of an electron-hole pair (or exciton). The absorption may have an increased probability at higher energies (i.e., shorter wavelengths) and result in a broadband absorption spectrum, in marked contrast to standard fluorophores.
  • quantum dots For nanocrystals smaller than the so-called Bohr exciton radius (a few nanometers), energy levels may be quantized, with values directly related to the qdot size (an effect called quantum confinement, hence the name "quantum dots").
  • Quantum dots The radiative recombination of an exciton (characterized by a long lifetime, >10 ns) may lead to the emission of a photon in a narrow, symmetric energy band.
  • the long fluorescence lifetime of qdots may enable the use of time-gated detection to separate their signal from that of shorter lived species (such as background autofluorescence encountered in cells).
  • Single qdots can be observed and tracked over an extended period of time with, for example, confocal microscopy, total internal reflection microscopy, or basic wide-field epifluorescence microscopy. Fluorescence correlation spectroscopy may allow determination of the brightness per particle and also provide a measurement of the average qdot size. Qdots can also be used as probes for two-photon confocal microscopy because they are characterized by a very large absorption cross section. They can be used simultaneously with standard dyes. Qdots have a potential as customizable donors of a fluorescence resonance energy transfer (FRET) pair.
  • FRET fluorescence resonance energy transfer
  • a single recognition moiety can be grafted to the qdot (e.g., DNA oligonucleotide or aptamer, antibody, antibody fragment, antigen-binding polypeptide, etc.) or, used as the qdot solubilization ligand.
  • Qdot ligands containing either an amine or a carboxyl group may offer a possibility of cross-linking molecules containing a thiol group or an N-hydroxysuccinimyl ester moiety by means of standard bioconjugation reactions.
  • streptavidin-coated qdots can be used in combination with biotinylated proteins or antibodies.
  • a three-layer approach such as, using (i) an antibody against a specific target, (ii) a biotinylated secondary antibody against the first, and (iii) a streptavidin-coated qdot can allow qdot labeling of non-natural amino acids, non-natural amino acid polypeptides, modified non-natural amino acid polypeptides and fragments thereof, as disclosed herein.
  • a number of potential surface attachment groups can be used to "graft" different functionalities to individual qdots, resulting in multipotent probes.
  • qdots can be equipped with a membrane-crossing or cell-internalization capability, and/or an enzymatic function.
  • Peptides can be customized, and with a choice of sequence, a single-step surfactant exchange can yield necessary functions: (i) protect the core/shell structure and maintain the original qdot photophysics, (ii) solubilize qdots, (iii) provide a biological interface, and (iv) allow the incorporation of multiple functions.
  • the resulting particles can have colloidal properties, photophysics, and biocompatibility, and this "peptide toolkit" can be tailored to provide additional functionalities. Such functionalities can be improved by molecular evolution.
  • Live-cell experiments such as, whole-cell labeling, labeling of membrane-bound proteins, and cytoplasmic or nuclear target labeling can be used for cell or pathogen detection, cell tracking, and cell lineage studies. This can be achieved without any functionalization through microinjection, electroporation, or phagocytosis of qdots.
  • Different types of functionalization can be explored as a way to target qdots to cell surface proteins.
  • Some examples include streptavidin, secondary, or primary antibodies, receptor ligands such as epidermal growth factor (EGF) or serotonin, recognition peptides, and affinity pairs such as biotin-avidin after engineering of the target protein.
  • Another strategy may consist of cross-linking primary antibodies to qdots.
  • Some proteins can be recognized by peptides, so peptides can be used for qdot functionalization.
  • Microinjection can allow the delivery of qdots functionalized with the appropriate targeting peptide sequence to mitochondria or the cell nucleus. The long-term stability and brightness of qdots make them a candidate for live animal targeting and imaging.
  • new compositions could entail qdots with properties such as (i) sensitivity to electric or magnetic fields; (ii) narrower fluorescence emission and longer lifetimes (using lanthanide-doped qdots); (iii) smaller sizes and extension to the NIR spectrum, as demonstrated by ternary alloys; (iv) end-specific functionalizations of nanorod qdots; (v) suppression of blinking and quantum yield enhancement; and (vi) built-in on-off switches or photoelectric biotransducers.
  • properties such as (i) sensitivity to electric or magnetic fields; (ii) narrower fluorescence emission and longer lifetimes (using lanthanide-doped qdots); (iii) smaller sizes and extension to the NIR spectrum, as demonstrated by ternary alloys; (iv) end-specific functionalizations of nanorod qdots; (v) suppression of blinking and quantum yield enhancement; and (vi) built-in on-off switches or photoelectric biotransduc
  • Biotransducer light-excited qdots could transfer their charge to bound enzymes functioning as electron or hole acceptors, enabling their control by light activation. Reciprocally, qdots could be lit up by electron or hole donor enzymes through chemiluminescence. Peptide coating of nano-materials can be a tool for imparting novel functions to the organic-inorganic interface.
  • the simultaneous engineering of the semiconductor's band gap (by rational design) with the peptide's redox potential (by molecular evolution) could be used to optimize qdot compositions and peptide sequences for binding and desired optical, electronic, magnetic, and chemical properties.
  • different shapes, end specificities, and compositions can lead to more complex bioinorganic architectures that could be exploited as an optoelectronic interface to the cellular machinery.
  • Qdots can be used as contrast reagents for functional imaging with a combination of MRI, PET, computed tomography, and IR fluorescence imaging (the latter by direct imaging through the epidermis or by a catheter-based confocal fiber microscope).
  • In vivo optical biopsy could confirm the pathology, and therapy could then be performed selectively, locally, and temporally by depositing energy (monochromatic x-rays for k-shell absorption or laser IR radiation) into the targeted qdots.
  • energy monochromatic x-rays for k-shell absorption or laser IR radiation
  • Monitoring the probes may consist of (1) a measurement system, and (2) an analysis software.
  • the measurement system may include all of the optics, electronics and the manner in which the sample is illuminated (e.g., light source selection), the mode of measurement (e.g., fluorescence or transmission), as well as the calibration best suited for extracting the desired results from the measurement.
  • the analysis software may include all of the software and mathematical algorithms necessary to analyze and display important results in a meaningful way.
  • the measurement can be carried out using virtually any optical system attached to the system, for example, an upright or inverted microscope, a fluorescence microscope, a macro lens, an endoscope and a fundus camera.
  • any standard experimental method can be used, including light transmission (bright field and dark field), autofluorescence and fluorescence of administered probes, etc. Fluorescence measurements can be made with any standard filter cube (consisting of a barrier filter, excitation filter and a dichroic mirror), or any customized filter cube for special applications, provided the emission spectra fall within the spectral range of the system sensitivity.
  • Radionuclide-labeled probes can be detected by PET or SPECT (single-photon emission tomography), probes emitting light (fluorescence, bioluminescence, or NIR emissions) can be detected by optical imaging, and radiowave emissions can be detected by MRI.
  • Small-animal devices can be used for radionuclide-based imaging (e.g., microSPECT and microPET), optical imaging of visible light (using sensitive, cooled charged-coupled device (CCD) cameras), and NIR emissions.
  • the combination of anatomical (microMRI and microCT) and molecular imaging techniques can help obtain molecular and functional information, and monitor specific molecular therapeutic efficacy.
  • Noninvasive reporter gene assays can be used for molecular-imaging studies of living animals. Radionuclide-labeled probes can be used to monitor, in living mice, the expression of reporter genes using the direct-binding FESP probe, or the herpes simplex virus type 1-thymidine kinase (HSV1-TK). HSV1-TK can be monitored with positron-labeled thymidine analogs. Like FDG, the indirect substrate probe for hexokinase, positron-labeled substrates for HSV1-TK can be retained in cells as a result of enzyme dependent phosphorylation. For optical-imaging assays, the light produced by the enzymes from their substrates can be monitored with sensitive CCD cameras. New reporter genes encoding fusion proteins that can be imaged with fluorescent, bioluminescent, or radionuclide probes can allow study of a single animal with a number of different imaging probes and instrumentation appropriate for distinct applications.
  • MicroPET instrumentation can provide better anatomic discrimination of functional assays: for example, pinpointing the locations of tumors within organs, determining the location of cell migration more accurately, etc.
  • Fluorescence-mediated tomography can improve the resolution and quantitation of optical imaging procedures.
  • Spectral-imaging technologies can discriminate emissions from multiple fluorescent probes, permitting simultaneous analysis of distinct optical probes and dramatically reducing background autofluorescence.
  • amino acids to be substituted in order to modulate activities or properties of the polypeptide may be done by site-directed mutagenesis.
  • Amino acids in the polypeptides and polypeptide libraries of the present invention that modulate function can be identified or modulated by substituting a non-naturally encoded amino acid in place of a natural amino acid at any or all positions of the polypeptide.
  • Naturally encoded amino acids may be substituted into a selected position of a polypeptide by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (See, e.g., Cunningham et al. 1989).
  • the alanine-scanning mutagenesis procedure introduces single alanine mutations at selected or every residue in the molecule.
  • a non-naturally encoded amino acid is substituted for a naturally encoded amino acid in the polypeptide chain.
  • the resulting mutant polypeptide molecules comprising a non-naturally encoded amino acid are then tested for biological activity using assays appropriate for measuring the function of the particular polypeptide or protein.
  • substitutions of non-naturally encoded charged amino acids or non-naturally encoded neutral amino acids for the naturally encoded charged and/or neutral amino acids. These substitutions may produce proteins with highly desirable improved or modulated characteristics, such as modulated receptor binding, modulated enzymatic activity, modulated antigen binding, or modulated aggregation or solubility.
  • This example describes conjugates that may be formed with non-natural amino acid polypeptides.
  • Molecules may be directly bonded to one or more non-natural amino acids in a polypeptide or may be attached via a linker, polymer, water soluble polymer, or biologically active molecule.
  • FIG. 9 presents non-limiting examples of molecules that are site specifically attached to polypeptides via a reaction that forms an oxime bond between the carbonyl of a non-natural amino acid incorporated into a polypeptide and the hydroxylamine of the molecule.
  • Molecules including but not limited to, fluorphores, biotin, and chelators may be attached to non-natural amino acid polypeptides.
  • FIG. 10 shows an example of a purification method for a non-natural amino acid polypeptide utilizing a resin that reacts with the non-natural amino acid.
  • a covalent linkage is formed between a chemically specific affinity tag on the resin and a non-natural amino acid present in the protein.
  • linkages are stable under a broad range of pH and purification conditions.
  • the separation step may be performed in alternate modes, including but not limited to a bath mode, enabling the large-scale purifications.
  • the resin and the affinity tags are physically and chemically stable, and thus, can be reused to reduce the cost of protein purification upon scale-up.
  • the separation can be performed in conjunction with conjugation of the polypeptide to molecules including but not limited to, PEG.
  • This "one-pot" method further simplifies the conjugation process and reduces the cost of production of proteins, including but not limited to target therapeutic proteins ( Figure 11 ).
  • Other molecules that can be conjugated include but are not limited to fluorphores.
  • Resins or other materials for purification can be selected and functionalized according to the non-natural amino acid present in the polypeptide.
  • Figure 12 shows an example of resin selection and functionalization.
  • Resins or other materials for purification can be functionalized differently depending on the non-natural amino acid in the polypeptide.
  • FIG. 13 shows an example of affinity purification of a non-natural amino acid polypeptide using hydroxylamine resin.
  • FIG. 14 shows an example of purification of a non-natural amino acid polypeptide using an aldehyde resin. Non-limiting examples of hydroxylamine and aldehyde resins are shown.
  • one or more steps of the purification process modify one or more non-natural amino acids present in the polypeptide to one or more natural amino acids.
  • FIG. 15 shows an example of purification of native proteins from a non-natural amino acid precursor.
  • the non-natural amino acid is converted to tyrosine after release from the resin used in the purification process.
  • FIG. 16 shows non-limiting examples of non-natural amino acids.
  • This example details cloning and expression of a hGH polypeptide including a non-naturally encoded amino acid in E. coli. This example also describes one method to assess the biological activity of modified hGH polypeptides.
  • An introduced translation system that comprises an orthogonal tRNA (O-tRNA) and an orthogonal aminoacyl tRNA synthetase (O-RS) is used to express hGH containing a non-naturally encoded amino acid.
  • the O-RS preferentially aminoacylates the O-tRNA with a non-naturally encoded amino acid.
  • the translation system inserts the non-naturally encoded amino acid into hGH, in response to an encoded selector codon.
  • Table 1 O-RS and O-tRNA sequences. SEQ ID NO:4 M.
  • E. coli with plasmids containing the modified hGH gene and the orthogonal aminoacyl tRNA synthetase/tRNA pair (specific for the desired non-naturally encoded amino acid) allows the site-specific incorporation of non-naturally encoded amino acid into the hGH polypeptide.
  • the transformed E. coli grown at 37° C in media containing between 0.01 - 100 mM of the particular non-naturally encoded amino acid, expresses modified hGH with high fidelity and efficiency.
  • the His-tagged hGH containing a non-naturally encoded amino acid is produced by the E. coli host cells as inclusion bodies or aggregates.
  • the aggregates are solubilized and affinity purified under denaturing conditions in 6M guanidine HCl. Refolding is performed by dialysis at 4°C overnight in 50mM TRIS-HCl, pH 8.0, 40 ⁇ M CuSO 4 , and 2% (w/v) Sarkosyl. The material is then dialyzed against 20mM TRIS-HCl, pH 8.0, 100mM NaCl, 2mM CaCl 2 , followed by removal of the His-tag. See Boissel et al., (1993) 268:15983-93. Methods for purification of hGH are well known in the art and are confirmed by SDS-PAGE, Western Blot analyses, or electrospray-ionization ion trap mass spectrometry and the like.
  • the His-tagged mutant hGH proteins were purified using the ProBond Nickel-Chelating Resin (Invitrogen, Carlsbad, CA) via the standard His-tagged protein purification procedures provided by the manufacturer, followed by an anion exchange column prior to loading on the gel.
  • an assay measuring a downstream marker of hGH's interaction with its receptor was used.
  • the interaction of hGH with its endogenously produced receptor leads to the tyrosine phosphorylation of a signal transducer and activator of transcription family member, STAT5, in the human IM-9 lymphocyte cell line.
  • Two forms of STAT5, STAT5A and STAT5B were identified from an IM-9 cDNA library.
  • IM-9 cells were stimulated with hGH polypeptides of the present invention.
  • the human IM-9 lymphocytes were purchased from ATCC (Manassas, VA) and grown in RPMI 1640 supplemented with sodium pyruvate, penicillin, streptomycin (Invitrogen, Carlsbad, San Diego) and 10% heat inactivated fetal calf serum (Hyclone, Logan, UT).
  • the IM-9 cells were starved overnight in assay media (phenol-red free RPMI, 10mM Hepes, 1% heat inactivated charcoal/dextran treated FBS, sodium pyruvate, penicillin and streptomycin) before stimulation with a 12-point dose range of hGH polypeptides for 10 min at 37°C.
  • Stimulated cells were fixed with 1% formaldehyde before permeabilization with 90% ice-cold methanol for 1 hour on ice.
  • the level of STAT5 phosphorylation was detected by intra-cellular staining with a primary phospho-STAT5 antibody (Cell Signaling Technology, Beverly, MA) at room temperature for 30 min followed by a PE-conjugated secondary antibody.
  • Sample acquisition was performed on the FACS Array with acquired data analyzed on the Flowjo software (Tree Star Inc., Ashland, OR).
  • EC 50 values were derived from dose response curves plotted with mean fluorescent intensity (MFI) against protein concentration utilizing SigmaPlot.
  • Table 2 summarizes the IM-9 data generated with mutant hGH polypeptides.
  • Various hGH polypeptides with a non-natural amino acid substitution at different positions were tested with human IM-9 cells as described. Substitutions shown were made with p-acetyl phenylalanine at the positions indicated. The same assay was used to assess biological activity of hGH polypeptides comprising a non-natural amino acid that is PEGylated. From the data shown in the table, it is apparent that there are differences in receptor binding activity depending upon the position in which the non-naturally encoded amino acid was substituted for a naturally encoded amino acid.
  • This example details cloning and expression of a modified hIFN polypeptide in E. coli.
  • This example demonstrates how a hIFN polypeptide including a non-naturally encoded amino acid can be expressed in E. coli. See Nagata et. al., Nature, vol. 284, 316-320 (1980 ) and U.S. Patent No. 4,364,863 .
  • cDNA encoding the full length hIFN and the mature form of hIFN lacking the N-terminal signal sequence are shown in SEQ ID NO: 23 and SEQ ID NO: 24, respectively.
  • the full length and mature hIFN encoding cDNA is inserted into the pBAD HISc, pET20b, and pET19b expression vectors following optimization of the sequence for cloning and expression without altering amino acid sequence.
  • An introduced translation system that comprises an orthogonal tRNA (O-tRNA) and an orthogonal aminoacyl tRNA synthetase (O-RS) is used to express hGH containing a non-naturally encoded amino acid.
  • the O-RS preferentially aminoacylates the O-tRNA with a non-naturally encoded amino acid.
  • the translation system inserts the non-naturally encoded amino acid into hGH, in response to an encoded selector codon.
  • O-RS and O-tRNA sequences suitable for use with Interferon expression include those shown in Example 3.
  • the transformation of E. coli with plasmids containing the modified hIFN gene and the orthogonal aminoacyl tRNA synthetase/tRNA pair allows the site-specific incorporation of non-naturally encoded amino acid into the hIFN polypeptide.
  • the transformed E. coli grown at 37° C in media containing between 0.01 - 100 mM of the particular non-naturally encoded amino acid, expresses modified hIFN with high fidelity and efficiency.
  • the His-tagged hIFN containing a non-naturally encoded amino acid is produced by the E. coli host cells and are affinity purified. Methods for purification of hIFN are well known in the art and are confirmed by SDS-PAGE, Western Blot analyses, or electrospray-ionization ion trap mass spectrometry and the like.
  • the hIFN receptor was prepared as described in U.S. Patents No. 6,566,132 ; 5,889,151 ; 5,861,258 ; 5,731,169 ; 5,578,707 .
  • a non-PEGylated polypeptide comprising a non-natural amino acid the affinity of the hormone for its receptor was measured by using a BIAcoreTM biosensor (Pharmacia) technique, which is known in the art.
  • BIAcore biosensor assays were used to measure the binding characteristics of hIFN molecules that comprised a non-naturally encoded amino acid substituted at the positions shown in Table 3, along with the receptor binding data.
  • Conjugates and complexes between proteins and oligonucleotides have wide applications in diagnosis and therapeutic, such as immunoPCR, gene therapeutic and more recently targeted delivery of RNAi.
  • Site-specific conjugation enables production of specifically designed molecules and nano structures that have novel functions.
  • the site-specific conjugations have been achieved mainly through maleimide chemistry, in which an engineered protein surface cysteine selectively reacts with maleimide to form a thioether.
  • the development of site-specific incorporation of unnatural amino acids into polypeptides has enabled a large array of chemistries for conjugation of molecules to proteins.
  • Over 30 non-naturally encoded amino acids have been incorporated site-specifically into proteins.
  • oligo nucleotides were conjugated to proteins site-specifically.
  • the conjugated proteins were assembled in one dimension in a defined manner.
  • Protein used in this experiment was human growth hormone Y35 mutant, in which the tyrosine 35 was replaced by the non-naturally encoded amino acid 9.2 (Scheme 1).
  • the single strand DNAs were stored as 25 mM solutions in water at -80°C.
  • the sequence of ssDNA FTam27 is 5'-CAG CCA GCG TGC ACG (SEQ ID NO:21).
  • the 5' of FTam27 was modified with hydrazide.
  • FTam28-d1 5'- CGT GCA CGC TGG CTG CGT GCA CGC TGG CTG (SEQ ID NO:21); FTam-d2: 5'- CGT GCA CGC TGG CTG T CGT GCA CGC TGG CTG (SEQ ID NO:22); FTam28-d3: 5'-CGT GCA CGC TGG CTG TT CGT GCA CGC TGG CTG: FTam28-t1 (SEQ ID NO:23); 5'- CGT GCA CGC TGG CTG CGT GCA CGC TGG CTG CGT GCA CGC TGG CTG (SEQ ID NO:24); FTam28-t2: 5'-CGT GCA CGC TGG CTG T CGT GCA CGC TGG T CTG CGT GCA CGC TGG CTG (SEQ ID NO:25); FTam28-t3: 5'-CGT GCA CGC TGG CTG TT CGT GCA CGC TGG CTG (SEQ
  • Protein (1mg) was buffer exchanged into reaction buffer (150 mM NaCl, 20 mM NaOAc, 400 mM Arg, 5 mM EDTA, pH 4.0) using PD 10 gel filtration columns.
  • the protein solution was concentrated to 90 ⁇ l using 10 kD MWCO CENTROCON (Vivascience).
  • the ssDNA solution was added slowly into the protein solution. Precipitation appeared initially, but dissolved. 20 hours after incubation at 28 °C, 5 mM sodium cyanoboronhydride was added. The reaction mixture was incubated for another 20 hours and subjected to analysis and purification.
  • a 1 ml phenyl HIC column was employed for the FPLC purification of the conjugate.
  • Buffer A 2 M NaCl, 10 mM Tris.HCl, pH 7.0
  • Buffer B 10 mM Tris.HCl, pH 7.0.
  • the gradient used in the purification was: 10 column volume (CV) 0% B, 5 CV to 50% B, hold at 50% B for 5 CV, then 30 CV to 100% B.
  • Purified conjugate was concentrated, buffer-exchanged to storage buffer (200 mM, NaCl, 50 mM Tris.HCl, 1 mM EDTA, pH 8.0) and subjected to PAGE analysis using 4-12% SDS gel, at 200 V in MES buffer.
  • Non-naturally encoded amino acid 9.2 which has a 1,3 diketone moiety, was incorporated into human growth hormone (hGH) at amino acid position 35, and used as a handle for the conjugation with a 15 mer single strand DNA, FTam27, modified at the 5' with hydrazide functional group (Scheme 1).
  • hGH human growth hormone
  • Scheme 1 This conjugation resulted in a hydrazone initially, which is further reduced with sodium cyanoborohydride to give an irreversible covalent linkage.
  • a 70% yield was obtained ( Figure 17 ).
  • the conjugate was purified to about 90% pure using HIC column and subject to hybridization.
  • the conjugate was designed to hybridize with ssDNAs that have two (d) or three (t) tandem complementary sequence (FTam28) repeats with zero(1), one(2) and two(3) bases T between them as spacers ( Figure 18 ).
  • FTam28 tandem complementary sequence
  • 5 ⁇ l of hGH-ssDNA conjugate was mixed with a series concentration of FTam28-d3, a single strand DNA that has two repeating sequences complementary to FTam27 and two T bases as a spacer between them.
  • the result was analyzed with 14% native glycine gel electrophoresis, 125 V, 3 hr at 4 °C ( Figure 19 ).
  • hybrid dimer there was a substantial amount of hybrid dimer present when 80 pico mole of hGH-ssDNA conjugate was mixed with 10 equivalents of FTam28-d3 (lane 3). This indicated that the hybrid dimer was more stable than the hybrid monomer thermodynamically.
  • single strand DNA was conjugated to the protein surface site-specifically.
  • This single strand DNA-protein conjugate can be used to assemble protein 1-D structures highly efficiently using DNA as a template.
  • Site-specific oligonucleotide conjugation can also be used to assemble well defined 3-D structures creating novel nano structures with novel functions.
  • the protein-oligo nucleotide conjugation technology may be applied to create protein drug "plug and play" libraries. In this case, the oligonucleotide may be used as both a linkage and a "name tag" to encode the individual small molecule and/or protein.
  • the protein-oligo nucleotide conjugate may be used in immunoPCR for diagnostic applications. This technology can also be used to create protein RNA or PNA conjugates which can be used in targeted RNAi therapeutics.

Claims (4)

  1. Composition comprenant un polypeptide lié de façon covalente à une molécule d'acide nucléique à une ou plusieurs positions d'acide aminé spécifiques dudit polypeptide, dans laquelle ladite molécule d'acide nucléique est liée de façon covalente à une chaîne latérale d'acide aminé d'un acide aminé non naturellement codé dudit polypeptide.
  2. Composition selon la revendication 1, dans laquelle l'acide nucléique est un ADN.
  3. Composition selon la revendication 2, dans laquelle l'ADN est un ADN monocaténaire (ADNsb).
  4. Composition selon l'une quelconque des revendications 1 à 3, dans laquelle le polypeptide est un anticorps.
EP10191428.1A 2005-11-16 2006-11-16 Procédés et compositions comprenant des acides aminés non naturels Active EP2339014B1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US73785505P 2005-11-16 2005-11-16
EP06837913A EP1951890A4 (fr) 2005-11-16 2006-11-16 Procedes et compositions comprenant des acides amines non naturels

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
EP06837913.0 Division 2006-11-16
EP06837913A Division EP1951890A4 (fr) 2005-11-16 2006-11-16 Procedes et compositions comprenant des acides amines non naturels

Publications (2)

Publication Number Publication Date
EP2339014A1 EP2339014A1 (fr) 2011-06-29
EP2339014B1 true EP2339014B1 (fr) 2015-05-27

Family

ID=38049316

Family Applications (2)

Application Number Title Priority Date Filing Date
EP06837913A Withdrawn EP1951890A4 (fr) 2005-11-16 2006-11-16 Procedes et compositions comprenant des acides amines non naturels
EP10191428.1A Active EP2339014B1 (fr) 2005-11-16 2006-11-16 Procédés et compositions comprenant des acides aminés non naturels

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP06837913A Withdrawn EP1951890A4 (fr) 2005-11-16 2006-11-16 Procedes et compositions comprenant des acides amines non naturels

Country Status (12)

Country Link
US (2) US20090018029A1 (fr)
EP (2) EP1951890A4 (fr)
JP (1) JP2009520949A (fr)
KR (1) KR20080079643A (fr)
CN (2) CN101454461A (fr)
AU (1) AU2006315347A1 (fr)
CA (1) CA2626522A1 (fr)
DK (1) DK2339014T3 (fr)
ES (1) ES2547554T3 (fr)
IL (3) IL190748A (fr)
PT (1) PT2339014E (fr)
WO (1) WO2007059312A2 (fr)

Families Citing this family (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009046445A1 (fr) * 2007-10-04 2009-04-09 Halcyon Molecular Séquençage de polymères d'acides nucléiques par microscopie électronique
JP2011503548A (ja) * 2007-11-02 2011-01-27 バイオトローブ, インコーポレイテッド 質量分析装置をクロマトグラフィシステムに連結するための装置および方法
JP5260976B2 (ja) * 2008-02-08 2013-08-14 東ソー株式会社 血液凝固第viii因子c2ドメインタンパク質の製造方法
US8940506B2 (en) 2008-03-21 2015-01-27 The Regents Of The University Of California High-sensitive fluorescent energy transfer assay using fluoresent amino acids and fluorescent proteins
EP2376647B1 (fr) * 2009-01-12 2014-03-05 Sutro Biopharma, Inc. Système de mono-chargement pour l'introduction sélective d'acides aminés non natifs dans des protéines à l'aide d'un système de synthèse de protéines in vitro
US9238878B2 (en) 2009-02-17 2016-01-19 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
CA2762277C (fr) * 2009-05-31 2018-08-14 Dh Technologies Development Pte. Ltd. Analyse specifique d'analytes de cetone et d'aldehyde au moyen de composes reactifs, de strategies d'etiquetage et de flux de travail de spectrometrie de masse
MX349301B (es) * 2009-12-21 2017-07-21 Ambrx Inc Polipéptidos de somatotropina bovina modificados y sus usos.
BR112012015597A2 (pt) * 2009-12-21 2017-01-31 Ambrx Inc peptídeos de somatotropina suínos modificados e seus usos
WO2011119771A2 (fr) * 2010-03-23 2011-09-29 The Salk Institute For Biological Studies Procédés et compositions pour détecter des modifications de protéine
HUE045845T2 (hu) 2010-08-17 2021-12-28 Ambrx Inc Módosított relaxin polipeptidek és felhasználásuk
US9567386B2 (en) 2010-08-17 2017-02-14 Ambrx, Inc. Therapeutic uses of modified relaxin polypeptides
DE102010064392A1 (de) * 2010-10-29 2012-05-03 Endress + Hauser Conducta Gesellschaft für Mess- und Regeltechnik mbH + Co. KG Verfahren zur Bestimmung eines Analytgehalts einer Flüssigkeitsprobe mittels eines Bioanalysators
RU2606016C2 (ru) 2011-01-14 2017-01-10 Редвуд Байосайнс, Инк. Меченые альдегидом полипептиды иммуноглобулина и способы их применения
JP5066615B2 (ja) 2011-01-31 2012-11-07 株式会社日立製作所 フェニルボロン酸基と特異的に結合するオリゴペプチド配列
EP2841056A4 (fr) 2012-04-23 2015-09-16 Massachusetts Inst Technology Particules enrobées couche par couche stables
JP6110611B2 (ja) * 2012-07-18 2017-04-05 株式会社東芝 アミンの回収方法および分析方法
WO2014059269A2 (fr) * 2012-10-12 2014-04-17 Massachusetts Institute Of Technology Compositions multicouches, dispositifs revêtus et leur utilisation
US10533947B2 (en) 2012-12-11 2020-01-14 Colin R Zamecnik Encapsulated dye coated noble metal nanoparticles with increased surface enhanced Raman scattering properties as contrast agents
CN103933575B (zh) 2013-01-23 2017-09-29 上海新理念生物医药科技有限公司 一种三齿型连接子及其应用
WO2014153164A1 (fr) 2013-03-14 2014-09-25 The California Institute For Biomedical Research Conjugués d'anticorps et d'agent de ciblage et leurs utilisations
KR102339240B1 (ko) 2013-10-15 2021-12-15 더 스크립스 리서치 인스티튜트 펩타이드 키메라 항원 수용체 t 세포 스위치 및 이의 용도
CN104573297B (zh) * 2013-10-24 2017-11-24 中国石油化工股份有限公司 一种确定表面催化反应路径的方法
WO2015109126A1 (fr) * 2014-01-15 2015-07-23 Caliper Life Sciences, Inc. Procédé et système d'immunodosage microfluidique au moyen de billes magnétiques
WO2016044328A1 (fr) * 2014-09-18 2016-03-24 The Regents Of The University Of California Analyse phénotypique de molécule unique
CN104697604B (zh) * 2015-03-13 2018-03-09 湖南菲尔斯特传感器有限公司 一种可现场校准的电容式液位传感器
WO2016154621A1 (fr) 2015-03-26 2016-09-29 The California Institute For Biomedical Research Récepteurs chimériques non-scfv commutables, commutateurs, et leurs utilisations
US11091546B2 (en) 2015-04-15 2021-08-17 The Scripps Research Institute Optimized PNE-based chimeric receptor T cell switches and uses thereof
US10775322B2 (en) 2015-06-16 2020-09-15 Arizona Board Of Regents On Behalf Of Arizona State University Inert crystal delivery medium for serial femtosecond crystallography
CN113350518A (zh) 2015-07-12 2021-09-07 杭州多禧生物科技有限公司 与细胞结合分子的共轭偶联的桥连接体
US9839687B2 (en) 2015-07-15 2017-12-12 Suzhou M-Conj Biotech Co., Ltd. Acetylenedicarboxyl linkers and their uses in specific conjugation of a cell-binding molecule
CN106932375A (zh) * 2015-12-31 2017-07-07 北京大学 蛋白质构象变化的生物正交拉曼原位检测方法
US20190079081A1 (en) * 2016-04-06 2019-03-14 Konica Minolta, Inc. Fluorescent immunostaining method
CA3019398A1 (fr) 2016-04-26 2017-11-02 R.P. Scherer Technologies, Llc Conjugues d'anticorps et methodes de fabrication et d'utilisation de ceux-ci
AU2017345479B2 (en) 2016-10-19 2024-03-21 The Scripps Research Institute Chimeric antigen receptor effector cell switches with humanized targeting moieties and/or optimized chimeric antigen receptor interacting domains and uses thereof
NZ752394A (en) 2016-11-14 2021-07-30 Hangzhou Dac Biotech Co Ltd Conjugation linkers, cell binding molecule-drug conjugates containing the likers, methods of making and uses such conjugates with the linkers
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
WO2018119003A1 (fr) * 2016-12-19 2018-06-28 The Regents Of The University Of California Biocapteur composite à virus
CA3052639A1 (fr) 2017-02-08 2018-08-16 Bristol-Myers Squibb Company Polypeptides de relaxine modifies comprenant un activateur pharmacocinetique et leurs utilisations
CN117286182A (zh) * 2017-06-02 2023-12-26 Ambrx公司 促进含非天然氨基酸的蛋白质产生的方法和组合物
US11419947B2 (en) 2017-10-30 2022-08-23 Massachusetts Institute Of Technology Layer-by-layer nanoparticles for cytokine therapy in cancer treatment
CN108519447A (zh) * 2018-04-02 2018-09-11 新疆出入境检验检疫局检验检疫技术中心 基于超高效液相色谱法测定干果中多种酸性染料的方法
EP3991752A4 (fr) 2019-06-29 2023-03-29 Hangzhou Dac Biotech Co., Ltd Conjugué molécule de liaison cellulaire-dérivé de tubulysine et méthode de préparation associée
CN110806448B (zh) * 2019-11-21 2022-03-22 北京市药品检验所 测定复方氨基酸注射液中游离氨的方法
CN114199844B (zh) * 2021-12-09 2024-02-09 吉林大学 一种金纳米簇及其在制备检测碱性磷酸酶荧光探针中的应用
CN115717130A (zh) * 2022-09-02 2023-02-28 凯莱英医药集团(天津)股份有限公司 氨酰-tRNA合酶突变体及烯基酪氨酰-tRNA的制备方法
WO2024077277A1 (fr) 2022-10-07 2024-04-11 Ambrx, Inc. Lieurs de médicaments et conjugués d'anticorps associés
CN116272921A (zh) * 2023-02-15 2023-06-23 青岛盛瀚色谱技术有限公司 一种单分散弱酸性阳离子色谱填料及其制备方法和应用

Family Cites Families (276)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4263428A (en) 1978-03-24 1981-04-21 The Regents Of The University Of California Bis-anthracycline nucleic acid function inhibitors and improved method for administering the same
US4289872A (en) 1979-04-06 1981-09-15 Allied Corporation Macromolecular highly branched homogeneous compound based on lysine units
US4898830A (en) 1979-07-05 1990-02-06 Genentech, Inc. Human growth hormone DNA
US4342832A (en) 1979-07-05 1982-08-03 Genentech, Inc. Method of constructing a replicable cloning vehicle having quasi-synthetic genes
ZA811368B (en) 1980-03-24 1982-04-28 Genentech Inc Bacterial polypedtide expression employing tryptophan promoter-operator
ATE12348T1 (de) 1980-11-10 1985-04-15 Gersonde Klaus Prof Dr Verfahren zur herstellung von lipid-vesikeln durch ultraschallbehandlung, anwendung des verfahrens und vorrichtung zur durchfuehrung des verfahrens.
US4364863A (en) 1980-12-29 1982-12-21 Schering Corporation Extraction of interferon from bacteria
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
FR2504010B1 (fr) 1981-04-15 1985-10-25 Sanofi Sa Medicaments anticancereux contenant la chaine a de la ricine associee a un anticorps antimelanome et procede pour leur preparation
US4551433A (en) 1981-05-18 1985-11-05 Genentech, Inc. Microbial hybrid promoters
JPS57206622A (en) 1981-06-10 1982-12-18 Ajinomoto Co Inc Blood substitute
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
NZ201705A (en) 1981-08-31 1986-03-14 Genentech Inc Recombinant dna method for production of hepatitis b surface antigen in yeast
JPS58118008A (ja) 1982-01-06 1983-07-13 Nec Corp デ−タ処理装置
EP0088046B1 (fr) 1982-02-17 1987-12-09 Ciba-Geigy Ag Lipides en phase aqueuse
US4671958A (en) 1982-03-09 1987-06-09 Cytogen Corporation Antibody conjugates for the delivery of compounds to target sites
DE3218121A1 (de) 1982-05-14 1983-11-17 Leskovar, Peter, Dr.-Ing., 8000 München Arzneimittel zur tumorbehandlung
EP0102324A3 (fr) 1982-07-29 1984-11-07 Ciba-Geigy Ag Lipides et composés tensio-actifs en phase aqueuse
US4512922A (en) 1982-12-22 1985-04-23 Genentech, Inc. Purification and activity assurance of precipitated heterologous proteins
US4511502A (en) 1982-12-22 1985-04-16 Genentech, Inc. Purification and activity assurance of precipitated heterologous proteins
US4511503A (en) 1982-12-22 1985-04-16 Genentech, Inc. Purification and activity assurance of precipitated heterologous proteins
US4820352A (en) 1983-01-10 1989-04-11 Bausch & Lomb Incorporated Cleaning and conditioning solutions for contact lenses and methods of use
CA1341116C (fr) 1983-02-22 2000-10-17 Rae Lyn Burke Systemes d'expression de la levure avec des vecteurs ayant des promoteurs de gapdh ou pyk, et synthese de proteine etrangere
US5089398A (en) 1983-02-22 1992-02-18 Chiron Corporation Enhanced yeast transcription employing hybrid GAPDH promoter region constructs
US4876197A (en) 1983-02-22 1989-10-24 Chiron Corporation Eukaryotic regulatable transcription
JPS59166086A (ja) 1983-03-09 1984-09-19 Teruhiko Beppu 新規な発現型プラスミドとそれらを用いて仔牛プロキモシン遺伝子を大腸菌内で発現させる方法
US4859600A (en) 1983-04-25 1989-08-22 Genentech, Inc. Recombinant procaryotic cell containing correctly processed human growth hormone
US4755465A (en) 1983-04-25 1988-07-05 Genentech, Inc. Secretion of correctly processed human growth hormone in E. coli and Pseudomonas
EP0127839B1 (fr) 1983-05-27 1992-07-15 THE TEXAS A&M UNIVERSITY SYSTEM Procédé pour la préparation d'un vecteur recombinant d'expression de baculovirus
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
JPS607934A (ja) 1983-06-29 1985-01-16 Dai Ichi Seiyaku Co Ltd リポソ−ムの製造方法
HUT35524A (en) 1983-08-02 1985-07-29 Hoechst Ag Process for preparing pharmaceutical compositions containing regulatory /regulative/ peptides providing for the retarded release of the active substance
US4689406A (en) 1983-08-10 1987-08-25 Amgen Enhancement of microbial expression of polypeptides
DE3486459D1 (de) 1983-09-26 1997-12-11 Udo Dr Med Ehrenfeld Mittel und Erzeugnis für die Diagnose und Therapie von Tumoren sowie zur Behandlung von Schwächen der zelligen und humoralen Immunabwehr
DE3474511D1 (en) 1983-11-01 1988-11-17 Terumo Corp Pharmaceutical composition containing urokinase
DK518384A (da) 1984-01-31 1985-07-01 Idaho Res Found Vektor til fremstilling af et gen-produkt i insektceller, fremgangsmaade til dens fremstilling samt dens anvendelse
EP0154316B1 (fr) 1984-03-06 1989-09-13 Takeda Chemical Industries, Ltd. Lymphokine chimiquement modifiée et son procédé de préparation
US4880734A (en) 1984-05-11 1989-11-14 Chiron Corporation Eukaryotic regulatable transcription
EP0164556B1 (fr) 1984-05-11 1994-03-02 Chiron Corporation Transcription augmentée dans les levures par usage d'une construction hybride de la région du promoteur
US4542225A (en) 1984-08-29 1985-09-17 Dana-Farber Cancer Institute, Inc. Acid-cleavable compound
US4738921A (en) 1984-09-27 1988-04-19 Eli Lilly And Company Derivative of the tryptophan operon for expression of fused gene products
US4659839A (en) 1984-10-10 1987-04-21 Mallinckrodt, Inc. Coupling agents for radiolabeled antibody fragments
US4837148A (en) 1984-10-30 1989-06-06 Phillips Petroleum Company Autonomous replication sequences for yeast strains of the genus pichia
GB8430252D0 (en) 1984-11-30 1985-01-09 Beecham Group Plc Compounds
ATE66469T1 (de) 1985-01-14 1991-09-15 Neorx Corp Metall-radionuklid markiertes protein fuer diagnose und therapie.
DE3675588D1 (de) 1985-06-19 1990-12-20 Ajinomoto Kk Haemoglobin, das an ein poly(alkenylenoxid) gebunden ist.
JP2524586B2 (ja) 1985-06-26 1996-08-14 シタス コーポレイション ポリマ−接合を利用する医薬組成物用蛋白質の可溶化
US4680338A (en) 1985-10-17 1987-07-14 Immunomedics, Inc. Bifunctional linker
US4699784A (en) 1986-02-25 1987-10-13 Center For Molecular Medicine & Immunology Tumoricidal methotrexate-antibody conjugate
GB8610600D0 (en) 1986-04-30 1986-06-04 Novo Industri As Transformation of trichoderma
JPS63123383A (ja) 1986-11-11 1988-05-27 Mitsubishi Kasei Corp ハイブリツドプロモ−タ−、発現調節dna配列および発現ベクタ−
US5186933A (en) 1986-12-30 1993-02-16 Baylor College Of Medicine Synthesis and immunogenicity of rotavirus genes using a baculovirus expression system
JPH02502876A (ja) 1987-03-16 1990-09-13 アメリカン バイオジェネティック サイエンシズ インク ワクチンと生物学的殺虫剤における組換えバクロウイルス閉塞体
CA1304020C (fr) 1987-03-23 1992-06-23 Meher H. Irani Haut niveau d'expression dans la levure
US5229490A (en) 1987-05-06 1993-07-20 The Rockefeller University Multiple antigen peptide system
CA1317244C (fr) 1987-07-24 1993-05-04 Ernest Seigo Kawasaki Production de formes biologiquement actives de csf a l'aide d'un systeme d'expression baculovirus (acnpv) - cellule d'insecte
WO1989001037A1 (fr) 1987-07-24 1989-02-09 Cetus Corporation Production de toxines de ricine dans un systeme d'expression de cellules d'insectes-baculovirus
US5080891A (en) 1987-08-03 1992-01-14 Ddi Pharmaceuticals, Inc. Conjugates of superoxide dismutase coupled to high molecular weight polyalkylene glycols
US4929555A (en) 1987-10-19 1990-05-29 Phillips Petroleum Company Pichia transformation
US4904584A (en) 1987-12-23 1990-02-27 Genetics Institute, Inc. Site-specific homogeneous modification of polypeptides
CA1340772C (fr) 1987-12-30 1999-09-28 Patricia Tekamp-Olson Expression et secretion de proteines heterologues dans une levure au moyen de sequences leader tronquees du facteur alpha
US4847325A (en) 1988-01-20 1989-07-11 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
CA1324969C (fr) 1988-05-06 1993-12-07 Jeffrey R. Shuster Expression a haut rendement de proteines dans la levure
US5674706A (en) 1988-05-06 1997-10-07 Chiron Corporation High level expression of proteins in yeast
FR2631974B1 (fr) 1988-05-31 1992-12-11 Agronomique Inst Nat Rech Baculovirus modifie, son procede de preparation et son application en tant que vecteur d'expression de genes
AU4197389A (en) 1988-08-05 1990-03-05 Mount Sinai School Of Medicine Of The City University Of New York, The In vivo infection of live insects with a recombinant baculovirus
GB8819453D0 (en) 1988-08-16 1988-09-21 Roy P Production of bluetongue virus non-structural proteins using baculovirus expression vector
NZ230425A (en) 1988-09-02 1992-07-28 Molecular Eng Ass Production of paramyxovirus fusion (f) protein using recombinant baculovirus expression vector
GB8824591D0 (en) 1988-10-20 1988-11-23 Royal Free Hosp School Med Fractionation process
US6780613B1 (en) 1988-10-28 2004-08-24 Genentech, Inc. Growth hormone variants
CA2001774C (fr) 1988-10-28 2001-10-16 James A. Wells Methode d'identification des domaines actifs et des residus d'acides amines dans les variants de polypeptides et d'hormones
WO1990005785A1 (fr) 1988-11-18 1990-05-31 The Regents Of The University Of California Procede d'incorporation sur un site specifique, d'aminoacides non naturels dans des proteines
DE68925966T2 (de) 1988-12-22 1996-08-29 Kirin Amgen Inc Chemisch modifizierte granulocytenkolonie erregender faktor
US4902502A (en) 1989-01-23 1990-02-20 Cetus Corporation Preparation of a polymer/interleukin-2 conjugate
WO1990010078A1 (fr) 1989-02-23 1990-09-07 University Of Ottawa Systeme ameliore d'expression de baculovirus capable de produire des proteines de genes etrangers en grande quantite
EP0460071A4 (en) 1989-02-24 1993-02-03 Cell Analysis Systems, Inc. Method and apparatus for determining a proliferation index of a cell sample
HUT64022A (en) 1989-04-19 1993-11-29 Enzon Inc Process for producing active polyalkileneoxide carbonates for the modification of polypeptides
US5324844A (en) 1989-04-19 1994-06-28 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5122614A (en) 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
CA2033070A1 (fr) 1989-05-17 1990-11-18 Lois K. Miller Vecteurs d'expression du baculovirus
EP0400472B1 (fr) 1989-05-27 1996-04-03 Sumitomo Pharmaceuticals Company, Limited Procédé pour la préparation de dérivés de polyéthylène glycol et protéine modifiée.
FR2649120B1 (fr) 1989-06-30 1994-01-28 Cayla Nouvelle souche et ses mutants de champignons filamenteux, procede de production de proteines recombinantes a l'aide de ladite souche et souches et proteines obtenues selon ce procede
IL91562A0 (en) 1989-09-07 1990-04-29 Yeda Res & Dev Interferon-gamma receptor fragment and its production
US5889151A (en) 1989-10-20 1999-03-30 Societe Leb-Tech Purified human alpha interferon receptor
US5312808A (en) 1989-11-22 1994-05-17 Enzon, Inc. Fractionation of polyalkylene oxide-conjugated hemoglobin solutions
US5162601A (en) 1989-11-22 1992-11-10 The Upjohn Company Plant potyvirus expression vector with a gene for protease
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5219564A (en) 1990-07-06 1993-06-15 Enzon, Inc. Poly(alkylene oxide) amino acid copolymers and drug carriers and charged copolymers based thereon
GB9206318D0 (en) 1992-03-24 1992-05-06 Cambridge Antibody Tech Binding substances
FR2664905B1 (fr) 1990-07-18 1994-08-12 Agronomique Inst Nat Rech Baculovirus modifie, son procede d'obtention, et vecteurs d'expression obtenus a partir dudit baculovirus.
WO1992001800A1 (fr) 1990-07-20 1992-02-06 Chiron Corporation Procede de transformation integrante de levure au moyen d'elements repetitifs disperses
WO1992002628A1 (fr) 1990-08-02 1992-02-20 Chiron Corporation Expression de glycoproteine-h de cytomegalovirus humain utilisant le systeme d'expression cellulaire de baculovirus-insectes
WO1992004363A1 (fr) 1990-09-04 1992-03-19 The Salk Institute Biotechnology/Industrial Associates, Inc. Production du facteur-1 de croissance similaire a l'insuline dans des cellules de levure methylotrophique
US5492821A (en) 1990-11-14 1996-02-20 Cargill, Inc. Stabilized polyacrylic saccharide protein conjugates
US5252714A (en) 1990-11-28 1993-10-12 The University Of Alabama In Huntsville Preparation and use of polyethylene glycol propionaldehyde
ES2113940T3 (es) 1990-12-03 1998-05-16 Genentech Inc Metodo de enriquecimiento para variantes de proteinas con propiedades de union alteradas.
US5231178A (en) 1991-01-16 1993-07-27 The Salk Institute Biotechnology/Industrial Associates, Inc. Method for the purification of intact, correctly-folded insulin-like growth factor-1
WO1992016555A1 (fr) 1991-03-18 1992-10-01 Enzon, Inc. Conjuges de polypeptides et de glycopolypeptides associes a des polymeres, contenant de l'hydrazine
AU1651992A (en) 1991-03-19 1992-10-21 United States of America, as represented by the Secretary, U.S. Department of Commerce, The Expression of influenza nucleoprotein antigens in baculovirus
US5595732A (en) 1991-03-25 1997-01-21 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US6126944A (en) 1991-04-26 2000-10-03 The United States Of America As Represented By The Department Of Health And Human Services Baculovirus expression vectors and recombinant antigens for detecting type-specific antibodies to herpes simplex virus
US6225447B1 (en) 1991-05-15 2001-05-01 Cambridge Antibody Technology Ltd. Methods for producing members of specific binding pairs
US5962255A (en) 1992-03-24 1999-10-05 Cambridge Antibody Technology Limited Methods for producing recombinant vectors
US6492160B1 (en) 1991-05-15 2002-12-10 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5281698A (en) 1991-07-23 1994-01-25 Cetus Oncology Corporation Preparation of an activated polymer ester for protein conjugation
US5290686A (en) 1991-07-31 1994-03-01 The United States Of America As Represented By The Department Of Health And Human Services Expression of influenza a M2 protein in baculovirus
US5866344A (en) 1991-11-15 1999-02-02 Board Of Regents, The University Of Texas System Antibody selection methods using cell surface expressed libraries
US5348867A (en) 1991-11-15 1994-09-20 George Georgiou Expression of proteins on bacterial surface
US5667988A (en) 1992-01-27 1997-09-16 The Scripps Research Institute Methods for producing antibody libraries using universal or randomized immunoglobulin light chains
IT1260468B (it) 1992-01-29 1996-04-09 Metodo per mantenere l'attivita' di enzimi proteolitici modificati con polietilenglicole
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
ATE156158T1 (de) 1992-04-14 1997-08-15 Cornell Res Foundation Inc Makromoleküle auf basis von dendritischen polymeren und verfahren zur herstellung
US5516657A (en) 1992-05-11 1996-05-14 Cambridge Biotech Corporation Baculovirus vectors for expression of secretory and membrane-bound proteins
ZA933926B (en) 1992-06-17 1994-01-03 Amgen Inc Polyoxymethylene-oxyethylene copolymers in conjuction with blomolecules
AU5006993A (en) 1992-08-21 1994-03-15 Enzon, Inc. Novel attachment of polyalkylene oxides to bio-effecting substances
US5382657A (en) 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
NZ250375A (en) 1992-12-09 1995-07-26 Ortho Pharma Corp Peg hydrazone and peg oxime linkage forming reagents and protein derivatives
US5298643A (en) 1992-12-22 1994-03-29 Enzon, Inc. Aryl imidate activated polyalkylene oxides
WO1994015625A1 (fr) 1993-01-15 1994-07-21 Enzon, Inc. Conjugues de polymere et de facteur viii
US5349001A (en) 1993-01-19 1994-09-20 Enzon, Inc. Cyclic imide thione activated polyalkylene oxides
US5321095A (en) 1993-02-02 1994-06-14 Enzon, Inc. Azlactone activated polyalkylene oxides
US5532142A (en) 1993-02-12 1996-07-02 Board Of Regents, The University Of Texas System Method of isolation and purification of fusion polypeptides
IL104734A0 (en) 1993-02-15 1993-06-10 Univ Bar Ilan Bioactive conjugates of cellulose with amino compounds
AU684510B2 (en) 1993-05-28 1997-12-18 Chiron Corporation Method for selection of biologically active peptide sequences
WO1994028024A1 (fr) 1993-06-01 1994-12-08 Enzon, Inc. Polymere modifie aux glucides presentant une activite erythropoïetique
WO1995000162A1 (fr) 1993-06-21 1995-01-05 Enzon, Inc. Synthese de peptides conjuges modifies, sur des sites specifiques
GB9317618D0 (en) 1993-08-24 1993-10-06 Royal Free Hosp School Med Polymer modifications
US5762939A (en) 1993-09-13 1998-06-09 Mg-Pmc, Llc Method for producing influenza hemagglutinin multivalent vaccines using baculovirus
US5643575A (en) 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
US5919455A (en) 1993-10-27 1999-07-06 Enzon, Inc. Non-antigenic branched polymer conjugates
US5491076A (en) 1993-11-01 1996-02-13 The Texas A&M University System Expression of foreign genes using a replicating polyprotein producing virus vector
US5605792A (en) 1993-11-04 1997-02-25 The Ohio State University Research Foundation Infectious bursal disease virus VP2 fusion protein expressed by baculovirus, use as diagnostic
PT730470E (pt) 1993-11-10 2002-08-30 Enzon Inc Conjugados melhorados de interferao-polimero
US5951974A (en) 1993-11-10 1999-09-14 Enzon, Inc. Interferon polymer conjugates
US5446090A (en) 1993-11-12 1995-08-29 Shearwater Polymers, Inc. Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
FR2715664B1 (fr) 1994-01-31 1996-04-12 Proteine Performance Sa Baculovirus recombinant et son utilisation pour la production d'anticorps monoclonaux.
JP3090586B2 (ja) 1994-03-15 2000-09-25 片倉工業株式会社 システインプロテアーゼ遺伝子欠損バキュロウイルスおよびその製造法並びにこれを利用する有用タンパク質の製造法
US5473034A (en) 1994-03-18 1995-12-05 Hyogo Prefectural Government Method for producing protein-synthetic polymer conjugate and said conjugate produced thereby
US5629384A (en) 1994-05-17 1997-05-13 Consiglio Nazionale Delle Ricerche Polymers of N-acryloylmorpholine activated at one end and conjugates with bioactive materials and surfaces
WO1995033490A1 (fr) 1994-06-02 1995-12-14 Enzon, Inc. Procede de solubilisation de substances pratiquement insolubles dans l'eau
US5635182A (en) 1994-06-16 1997-06-03 Genetics Institute, Inc. Method of detecting ligand interactions
US5730990A (en) 1994-06-24 1998-03-24 Enzon, Inc. Non-antigenic amine derived polymers and polymer conjugates
US6403375B1 (en) 1994-08-24 2002-06-11 Boyce Thompson Institute For Plant Research, Inc. Establishment of Trichoplusia ni cell lines in serum-free medium for recombinant protein and baculovirus production
US5688670A (en) 1994-09-01 1997-11-18 The General Hospital Corporation Self-modifying RNA molecules and methods of making
US5650234A (en) 1994-09-09 1997-07-22 Surface Engineering Technologies, Division Of Innerdyne, Inc. Electrophilic polyethylene oxides for the modification of polysaccharides, polypeptides (proteins) and surfaces
US5871986A (en) 1994-09-23 1999-02-16 The General Hospital Corporation Use of a baculovirus to express and exogenous gene in a mammalian cell
US5824784A (en) 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
WO1996041813A2 (fr) 1994-11-09 1996-12-27 Offord Robin E Polymeres fonctionnalises destines a une ligation dirigee
US5738846A (en) 1994-11-10 1998-04-14 Enzon, Inc. Interferon polymer conjugates and process for preparing the same
IL116085A (en) 1994-12-16 1999-12-31 Ortho Pharma Corp Spray dried erythropoietin
US5932462A (en) 1995-01-10 1999-08-03 Shearwater Polymers, Inc. Multiarmed, monofunctional, polymer for coupling to molecules and surfaces
AU720337B2 (en) 1995-02-17 2000-05-25 Stichting Dienst Landbouwkundig Onderzoek Production of biologically active recombinant bovine follicle stimulating hormone (REC bFSH) in the baculovirus expression system
FR2732035B1 (fr) 1995-03-23 1997-05-30 Agronomique Inst Nat Rech Procede de regulation de l'expression d'un gene dans un baculovirus, par un site de fixation d'un recepteur de l'acide retinoique, et vecteur pour la mise en oeuvre du dit procede
US6184344B1 (en) 1995-05-04 2001-02-06 The Scripps Research Institute Synthesis of proteins by native chemical ligation
US5702892A (en) 1995-05-09 1997-12-30 The United States Of America As Represented By The Department Of Health And Human Services Phage-display of immunoglobulin heavy chain libraries
NZ308772A (en) 1995-05-17 1999-04-29 Du Pont Recombinant baculovirus insecticides
US6475806B1 (en) 1995-06-07 2002-11-05 Praecis Pharmaceuticals, Inc. Anchor libraries and identification of peptide binding sequences
AU5893696A (en) 1995-06-07 1996-12-30 Novo Nordisk A/S Modification of polypeptides
US5672662A (en) 1995-07-07 1997-09-30 Shearwater Polymers, Inc. Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications
US5861279A (en) 1996-01-17 1999-01-19 Schering Corporation Baculovirus expression system for human interleukin 5 receptor and method of screening for interleukin 5 antagonists
AU1690697A (en) 1996-01-17 1997-08-11 Schering Corporation Baculovirus expression system for human interleukin 5 receptor and method of screening for interleukin antagonists
US5747639A (en) 1996-03-06 1998-05-05 Amgen Boulder Inc. Use of hydrophobic interaction chromatography to purify polyethylene glycols
TW517067B (en) 1996-05-31 2003-01-11 Hoffmann La Roche Interferon conjugates
US6083693A (en) 1996-06-14 2000-07-04 Curagen Corporation Identification and comparison of protein-protein interactions that occur in populations
CA2262994A1 (fr) 1996-08-02 1998-02-12 Ortho-Mcneil Pharmaceutical, Inc. Polypeptides possedant un polymere soluble dans l'eau a n-terminal lie par covalence simple
US5980948A (en) 1996-08-16 1999-11-09 Osteotech, Inc. Polyetherester copolymers as drug delivery matrices
US6214966B1 (en) 1996-09-26 2001-04-10 Shearwater Corporation Soluble, degradable poly(ethylene glycol) derivatives for controllable release of bound molecules into solution
US6083910A (en) 1996-12-13 2000-07-04 Chiron Corporation Therapeutic uses of resolved intact or clipped native-sequence PDGF-BB dimers
US6261804B1 (en) 1997-01-21 2001-07-17 The General Hospital Corporation Selection of proteins using RNA-protein fusions
WO1998031700A1 (fr) 1997-01-21 1998-07-23 The General Hospital Corporation Selection de proteines a l'aide de fusions arn-proteine
ATE200030T1 (de) 1997-01-29 2001-04-15 Polymasc Pharmaceuticals Plc Pegylationsverfahren
GB9703406D0 (en) 1997-02-19 1997-04-09 Chiron Spa Expression of heterologous proteins
JP4086325B2 (ja) 1997-04-23 2008-05-14 プリュックテュン,アンドレアス 標的分子と相互作用する(ポリ)ペプチドをコードする核酸分子の同定方法
AU7266898A (en) 1997-04-30 1998-11-24 Enzon, Inc. Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
US6180341B1 (en) 1997-05-01 2001-01-30 Board Of Regents, The Universiry Of Texas System In vitro scanning saturation mutagenesis of proteins
US5990237A (en) 1997-05-21 1999-11-23 Shearwater Polymers, Inc. Poly(ethylene glycol) aldehyde hydrates and related polymers and applications in modifying amines
ATE469166T1 (de) 1997-06-06 2010-06-15 Kyowa Hakko Kirin Co Ltd Chemisch modifizierte polypeptide
US5965393A (en) 1997-07-01 1999-10-12 National Institute Of Immunology Method for enhancing foreign gene expression in baculovirus expression vector system
NZ502375A (en) 1997-07-14 2001-11-30 Bolder Biotechnology Inc The addition of non-natural cysteine derivatives to cause the protein to act as antagonists of the GH family
GB9715660D0 (en) 1997-07-25 1997-10-01 Zeneca Ltd Proteins
EP1003889A1 (fr) 1997-08-05 2000-05-31 Chiron Corporation Nouvelles sequences de gene pichia pastoris et procedes d'utilisation de ces dernieres
DE19735593C2 (de) 1997-08-15 1999-08-26 Hepavec Ag Fuer Gentherapie Hüllprotein-modifizierter Baculovirus-Vektor für die Gentherapie
US6090584A (en) 1997-08-21 2000-07-18 University Technologies International Inc. Baculovirus artificial chromosomes and methods of use
US5989868A (en) 1997-09-12 1999-11-23 The Board Of Regents Of The University Of Oklahoma Fusion protein systems designed to increase soluble cytoplasmic expression of heterologous proteins in esherichia coli
AU9393398A (en) 1997-09-16 1999-04-05 Egea Biosciences, Inc. Method for the complete chemical synthesis and assembly of genes and genomes
AU752037B2 (en) 1997-09-26 2002-09-05 Uab Research Foundation Reduced antigenic cells and uses therefor
US6201072B1 (en) 1997-10-03 2001-03-13 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co- glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6004573A (en) 1997-10-03 1999-12-21 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6238865B1 (en) 1997-10-17 2001-05-29 Guangtian Chen Simple and efficient method to label and modify 3′-termini of RNA using DNA polymerase and a synthetic template with defined overhang nucleotides
DE19748489A1 (de) 1997-11-03 1999-05-06 Roche Diagnostics Gmbh Polyethylenglykol-derivatisierte Biomoleküle und deren Verwendung in heterogenen Nachweisverfahren
US6448369B1 (en) 1997-11-06 2002-09-10 Shearwater Corporation Heterobifunctional poly(ethylene glycol) derivatives and methods for their preparation
EP0924298A1 (fr) 1997-12-18 1999-06-23 Stichting Instituut voor Dierhouderij en Diergezondheid (ID-DLO) Expression de proteine dans des systèmes d'expression faisant appel aux baculovirus
US5985263A (en) 1997-12-19 1999-11-16 Enzon, Inc. Substantially pure histidine-linked protein polymer conjugates
US5981709A (en) 1997-12-19 1999-11-09 Enzon, Inc. α-interferon-polymer-conjugates having enhanced biological activity and methods of preparing the same
JP2002505110A (ja) 1998-03-04 2002-02-19 オニックス ファーマシューティカルズ,インコーポレイティド 遺伝物質の高生産性発現のためのバキュロウイルス発現系及び方法
ATE279430T1 (de) 1998-03-05 2004-10-15 Chiron Corp Verfahren zur verbesserung der serum halbwertszeit von biologisch aktiven molekülen
ES2307865T3 (es) 1998-03-12 2008-12-01 Nektar Therapeutics Al, Corporation Metodo para preparar conjugados polimericos.
KR100264953B1 (ko) 1998-04-03 2001-03-02 박현우 재조합 베큘로바이러스, 이의 제조방법 및 이를 포함하는 미생물 살충제
US6440695B1 (en) 1998-04-17 2002-08-27 Whitehead Institute For Biomedical Research Method for producing diverse libraries of encoded polypeptides
US6277588B1 (en) 1998-05-01 2001-08-21 Tel Aviv University Screening of cell populations
JP4240574B2 (ja) 1998-05-15 2009-03-18 三菱化学株式会社 タンパク質のラベル化組成物およびタンパク質のラベル化方法
US6451986B1 (en) 1998-06-22 2002-09-17 Immunex Corporation Site specific protein modification
US6168932B1 (en) 1998-07-13 2001-01-02 Parker Hughes Institute Recombinant DTctGMCSF fusion toxin in a baculovirus expression vector system
US6245528B1 (en) 1998-07-28 2001-06-12 Academia Sinica Latent baculovirus expression system
US6368825B1 (en) 1998-08-10 2002-04-09 Academia Sinica Baculovirus containing minimal CMV promoter
DE69941193D1 (de) 1998-08-17 2009-09-10 Bristol Myers Squibb Co Identifizierung von verbindung-protein wechselwirkungen mit bibliotheken von gekoppelten protein-nukleinsäure-molekülen
CN1330675A (zh) 1998-08-28 2002-01-09 格莱风科学公司 精确长度的聚酰胺链、其制备方法和其与蛋白质的缀合物
WO2000020032A1 (fr) 1998-10-06 2000-04-13 Trustees Of Dartmouth College ALLERGENE RECOMBINANT DU CHAT, Fel dI, EXPRIMES DANS BACULOVIRUS POUR DIAGNOSTIQUER OU TRAITER L'ALLERGIE AUX CHATS
US6420339B1 (en) 1998-10-14 2002-07-16 Amgen Inc. Site-directed dual pegylation of proteins for improved bioactivity and biocompatibility
CA2348822A1 (fr) 1998-10-30 2000-05-11 Novozymes A/S Proteines glycosylees a allergenicite reduite
WO2000032823A1 (fr) 1998-12-02 2000-06-08 Phylos, Inc. Fusions de proteine-adn et leurs utilisations
US6451346B1 (en) 1998-12-23 2002-09-17 Amgen Inc Biodegradable pH/thermosensitive hydrogels for sustained delivery of biologically active agents
US6281211B1 (en) 1999-02-04 2001-08-28 Euro-Celtique S.A. Substituted semicarbazides and the use thereof
US6342216B1 (en) 1999-03-17 2002-01-29 The Board Of Regents, The University Of Texas System Therapy of cancer by insect cells containing recombinant baculovirus encoding genes
US6994986B2 (en) 1999-03-17 2006-02-07 The Board Of Trustees Of The Leland Stanford University In vitro synthesis of polypeptides by optimizing amino acid metabolism
AU4325900A (en) 1999-03-17 2000-10-04 Board Of Trustees Of The Leland Stanford Junior University In vitro macromolecule biosynthesis methods using exogenous amino acids and a novel atp regeneration system
US6337191B1 (en) 1999-03-22 2002-01-08 The Board Of Trustees Of The Leland Stanford Junior University Vitro protein synthesis using glycolytic intermediates as an energy source
WO2000063268A1 (fr) 1999-04-16 2000-10-26 Wm. Marsh Rice University Poly(propylene fumarate) reticule avec du poly(ethylene glycol)
US6818112B2 (en) * 1999-04-20 2004-11-16 Target Discovery, Inc. Protein separation via multidimensional electrophoresis
EP1187626A4 (fr) 1999-06-01 2006-07-19 Compound Therapeutics Inc Procedes de production de conjugats de 5'-acide nucleique-proteine
CA2372795A1 (fr) 1999-07-12 2001-01-18 Robert G. Kuimelis Marquage de proteines c-terminales
US6261805B1 (en) 1999-07-15 2001-07-17 Boyce Thompson Institute For Plant Research, Inc. Sialyiation of N-linked glycoproteins in the baculovirus expression vector system
ES2383332T3 (es) 1999-07-27 2012-06-20 Bristol-Myers Squibb Company Procedimiento de ligación de aceptores de péptidos
WO2001016352A1 (fr) 1999-08-27 2001-03-08 Phylos, Inc. Methodes de codage et de tri de proteines traduites in vitro
US20020019061A1 (en) * 1999-08-31 2002-02-14 Hung-Sen Lai Purification and analysis of cyclic peptide libraries, and compositions thereof
AU2039501A (en) 1999-10-15 2001-04-23 Rockefeller University, The System for rapid generation of recombinant baculovirus-based expression vectors for silkworm larvae
US6232074B1 (en) 1999-12-10 2001-05-15 Complegen, Inc. Functional gene array in yeast
US6348558B1 (en) 1999-12-10 2002-02-19 Shearwater Corporation Hydrolytically degradable polymers and hydrogels made therefrom
MXPA02006216A (es) 1999-12-22 2004-02-26 Nektar Therapeutics Al Corp Derivados estericamente impedidos de polimeros solubles en agua.
WO2001045796A2 (fr) 1999-12-22 2001-06-28 Shearwater Corporation Procede de preparation d"esters de 1-benzotriazolyl carbonate de poly(ethylene glycol)
US6413507B1 (en) 1999-12-23 2002-07-02 Shearwater Corporation Hydrolytically degradable carbamate derivatives of poly (ethylene glycol)
US6646110B2 (en) 2000-01-10 2003-11-11 Maxygen Holdings Ltd. G-CSF polypeptides and conjugates
WO2001053539A1 (fr) 2000-01-24 2001-07-26 Phylos, Inc. Methodes diagnostiques multiplexees et sensibles destines a l'analyse des proteines
WO2001062827A2 (fr) 2000-02-22 2001-08-30 Shearwater Corporation Derives de polymere n-maleimidyle
AU2001257577A1 (en) 2000-02-28 2001-09-03 Shearwater Corporation Water-soluble polymer conjugates of artelinic acid
US6495337B1 (en) 2000-03-29 2002-12-17 Cytokinetics, Inc. High throughput sarcomeric assay
GB0012997D0 (en) 2000-05-26 2000-07-19 Eurogene Limited Gene delivery
US6586207B2 (en) 2000-05-26 2003-07-01 California Institute Of Technology Overexpression of aminoacyl-tRNA synthetases for efficient production of engineered proteins containing amino acid analogues
US6406863B1 (en) 2000-06-23 2002-06-18 Genetastix Corporation High throughput generation and screening of fully human antibody repertoire in yeast
US6410271B1 (en) 2000-06-23 2002-06-25 Genetastix Corporation Generation of highly diverse library of expression vectors via homologous recombination in yeast
US6410246B1 (en) 2000-06-23 2002-06-25 Genetastix Corporation Highly diverse library of yeast expression vectors
US6951947B2 (en) 2000-07-13 2005-10-04 The Scripps Research Institute Labeled peptides, proteins and antibodies and processes and intermediates useful for their preparation
JP2002030098A (ja) 2000-07-17 2002-01-29 Institute Of Immunology Co Ltd バキュロウィルスの発芽ウイルスからウイルスエンベロープを回収する方法
RU2003109746A (ru) 2000-09-08 2005-01-27 Грифон Терапьютикс, Инк. (Us) Синтетические белки, стимулирующие эритропоэз
US6610472B1 (en) 2000-10-31 2003-08-26 Genetastix Corporation Assembly and screening of highly complex and fully human antibody repertoire in yeast
US6436386B1 (en) 2000-11-14 2002-08-20 Shearwater Corporation Hydroxyapatite-targeting poly (ethylene glycol) and related polymers
TW593427B (en) 2000-12-18 2004-06-21 Nektar Therapeutics Al Corp Synthesis of high molecular weight non-peptidic polymer derivatives
TWI246524B (en) 2001-01-19 2006-01-01 Shearwater Corp Multi-arm block copolymers as drug delivery vehicles
JP2005502322A (ja) 2001-04-19 2005-01-27 ザ スクリップス リサーチ インスティテュート 非天然アミノ酸のインビボ組込み
US6566132B1 (en) 2001-04-26 2003-05-20 Isis Pharmaceuticals, Inc. Antisense modulation of Interferon gamma receptor 1 expression
US20050053973A1 (en) * 2001-04-26 2005-03-10 Avidia Research Institute Novel proteins with targeted binding
GB0113657D0 (en) 2001-06-05 2001-07-25 Geneprot Inc Improved native chemical ligation with three or more components
US20040138412A1 (en) 2001-09-07 2004-07-15 Paolo Botti Extended native chemical ligation
US6908963B2 (en) 2001-10-09 2005-06-21 Nektar Therapeutics Al, Corporation Thioester polymer derivatives and method of modifying the N-terminus of a polypeptide therewith
US7026440B2 (en) 2001-11-07 2006-04-11 Nektar Therapeutics Al, Corporation Branched polymers and their conjugates
CA2466746A1 (fr) 2001-11-14 2003-05-22 Geneprot, Inc. Ligation peptidique chimique avec au moins trois composants
US6716821B2 (en) 2001-12-21 2004-04-06 Immunogen Inc. Cytotoxic agents bearing a reactive polyethylene glycol moiety, cytotoxic conjugates comprising polyethylene glycol linking groups, and methods of making and using the same
US7622248B2 (en) 2002-02-15 2009-11-24 The Research Foundation Of State University Of New York Ribozymes with broad tRNA aminoacylation activity
WO2003089454A2 (fr) * 2002-04-19 2003-10-30 California Institute Of Technology Bibliotheque de molecules d'acides nucleiques et de peptides dont les peptides contiennent des residus d'acides amines non naturels, et methodes de production associee
EP2226316B1 (fr) 2002-05-30 2016-01-13 The Scripps Research Institute Ligation d'azides et d'acetylénes catalysée par le cuivre
EP1396498A1 (fr) * 2002-09-04 2004-03-10 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Paire de polypeptides avec une intéraction spécifique et forte
ATE445709T1 (de) 2002-10-16 2009-10-15 Scripps Research Inst Glycoproteinsynthese
MXPA05003983A (es) 2002-10-16 2005-06-22 Scripps Research Inst Incorporacion en sitio especifico de cetoaminoacidos en proteinas.
US20040171085A1 (en) * 2002-11-04 2004-09-02 Irm Llc Methods and compositions for treating neurodegenerative diseases
EP1583816A4 (fr) * 2002-12-22 2007-06-13 Scripps Research Inst Reseaux de proteines
US20040146855A1 (en) * 2003-01-27 2004-07-29 Marchessault Robert H. Formation of superparamagnetic particles
CN101223272B (zh) 2003-04-17 2013-04-03 斯克利普斯研究院 扩展真核生物遗传密码
AU2004267359B2 (en) 2003-07-07 2010-09-09 The Scripps Research Institute Compositions of orthogonal lysyl-tRNA and aminoacyl-tRNA synthetase pairs and uses thereof
EP1704242A4 (fr) 2003-07-07 2008-06-18 Scripps Research Inst COMPOSITIONS DE PAIRES ORTHOGONALES DE LEUCYL-tRNA ET AMINOACYL-tRNA-SYNTHETASE ET LEURS UTILISATIONS
US7527943B2 (en) 2003-07-07 2009-05-05 The Scripps Research Institute Compositions of orthogonal glutamyl-tRNA and aminoacyl-tRNA synthetase pairs and uses thereof
SG143252A1 (en) 2003-10-09 2008-06-27 Ambrx Inc Polymer derivatives
US7741071B2 (en) * 2003-12-18 2010-06-22 The Scripps Research Institute Selective incorporation of 5-hydroxytryptophan into proteins in mammalian cells
ATE428779T1 (de) * 2003-12-18 2009-05-15 Biomethodes Verfahren zur ortspezifischen massenmutagenese
AU2005211362B2 (en) 2004-02-02 2008-03-13 Ambrx, Inc. Modified human interferon polypeptides and their uses
MX2007000728A (es) * 2004-07-21 2007-03-15 Ambrx Inc Polipeptidos biosinteticos que utilizan amino acidos no naturalmente codificados.

Also Published As

Publication number Publication date
WO2007059312A2 (fr) 2007-05-24
WO2007059312A3 (fr) 2008-04-03
KR20080079643A (ko) 2008-09-01
IL190748A (en) 2013-05-30
IL225739A0 (en) 2013-06-27
EP1951890A2 (fr) 2008-08-06
JP2009520949A (ja) 2009-05-28
US9488660B2 (en) 2016-11-08
IL190748A0 (en) 2008-11-03
EP1951890A4 (fr) 2009-06-24
EP2339014A1 (fr) 2011-06-29
US20110144307A1 (en) 2011-06-16
PT2339014E (pt) 2015-10-13
DK2339014T3 (en) 2015-07-20
US20090018029A1 (en) 2009-01-15
IL225739A (en) 2017-10-31
CN106443006A (zh) 2017-02-22
CN101454461A (zh) 2009-06-10
AU2006315347A1 (en) 2007-05-24
ES2547554T3 (es) 2015-10-07
CA2626522A1 (fr) 2007-05-24

Similar Documents

Publication Publication Date Title
EP2339014B1 (fr) Procédés et compositions comprenant des acides aminés non naturels
Liu et al. Mass spectrometry-based protein footprinting for higher-order structure analysis: fundamentals and applications
KR101332875B1 (ko) 비-천연 아미노산 및 폴리펩티드를 함유하는 조성물,비-천연 아미노산 및 폴리펩티드를 포함하는 방법, 및비-천연 아미노산 및 폴리펩티드의 용도
US20210223252A1 (en) Methods for proteomic profiling using non-natural amino acids
Khidekel et al. Probing the dynamics of O-GlcNAc glycosylation in the brain using quantitative proteomics
KR20090102838A (ko) 페나진 및 퀴녹살린 치환된 아미노산 및 폴리펩티드
BRPI0516385B1 (pt) Composto ou sal do mesmo contendo aminoácidos não naturais e polipeptídeos, métodos para produção e derivatização de um polipeptídeo
US8945861B2 (en) Methods for isotopically labeling biomolecules using mammalian cell-free extracts
KR20080081013A (ko) 비천연 아미노산 및 폴리펩티드를 함유하는 조성물, 그와관련된 방법 및 그 용도
JP5565989B2 (ja) 新生タンパク質の検出、分析、及び分離方法
Li et al. Proteomic strategies for characterizing ubiquitin-like modifications
US8507648B2 (en) Detection of truncation mutations by mass spectrometry
Furuki et al. A novel rapid analysis using mass spectrometry to evaluate downstream refolding of recombinant human insulin‐like growth factor‐1 (mecasermin)
US20240036055A1 (en) Multiplexed DiLeu-Biotin-Azide (DBA) Tag Enabled Isobaric Tandem Orthogonal Proteolysis Activity-Based Protein Profiling (isoBOP-ABPP) Platform For High-Throughput Quantitative Pan-PTM Analysis
Matsumoto et al. Chemical genetic interaction linking eIF5A hypusination and mitochondrial integrity
Homan et al. Photoaffinity labelling with small molecules
You et al. Analysis of a macrophage carbamylated proteome reveals a function in post-translational modification crosstalk
Merz Development and Characterization of a FKBP51FK1 Affinity Matrix for Protein-Protein Interaction Studies
EP1944367A1 (fr) Technique de mesure du degré de polymérisation d'une proteine
Gilany et al. Mass spectrometry-based proteomics in the life sciences: a review
Aslebagh et al. Proteomics of the post-translational modifications: the knowns and the unknowns
Blake-Haskins et al. CHARACTERIZATION NEEDED PRIOR TO PROOF OF CONCEPT

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AC Divisional application: reference to earlier application

Ref document number: 1951890

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

17P Request for examination filed

Effective date: 20111220

17Q First examination report despatched

Effective date: 20120917

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20141209

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AC Divisional application: reference to earlier application

Ref document number: 1951890

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK RS

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 728898

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150615

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602006045569

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20150717

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: E. BLUM AND CO. AG PATENT- UND MARKENANWAELTE , CH

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2547554

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20151007

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20150827

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150827

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150927

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150828

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

Ref country code: RO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150527

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602006045569

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20160301

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151116

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20061116

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 728898

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150527

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150527

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: EE

Payment date: 20171222

Year of fee payment: 15

Ref country code: DK

Payment date: 20171110

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PT

Payment date: 20181112

Year of fee payment: 13

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20181130

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20181201

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 728898

Country of ref document: AT

Kind code of ref document: T

Effective date: 20181116

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20190410

Year of fee payment: 13

Ref country code: IE

Payment date: 20190410

Year of fee payment: 13

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181201

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20190410

Year of fee payment: 13

Ref country code: BE

Payment date: 20190417

Year of fee payment: 13

Ref country code: FR

Payment date: 20190410

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20190416

Year of fee payment: 13

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181116

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181116

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20181130

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20190410

Year of fee payment: 13

Ref country code: ES

Payment date: 20190730

Year of fee payment: 13

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602006045569

Country of ref document: DE

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200619

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191130

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191130

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20191130

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191117

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20191116

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200603

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191116

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191116

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191130

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20210527

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191117

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201130